$340K Collaborative Grant Funds Research on the Immune Response to Sarcomas

October 21, 2015 - The Liddy Shriver Sarcoma Initiative is pleased to announce the awarding of a $340,400 research grant for a collaborative study on the immune system and the potential role of immunotherapy in sarcomas. The grant brings together investigators from Austalia, Canada, France and the United States in a two-year study of two sarcoma subtypes.

Using medicine to help a patient's immune system fight cancer is an exciting area of research today. Despite the fact that cancer immunotherapy was named Science magazine’s 2013 advance of the year, few immunotherapy options exist for people with sarcomas. This study aims to reveal the immune system's role in osteosarcoma and undifferentiated pleomorphic sarcoma and to build a foundation for the use of immunotherapy in sarcoma treatment.

Osteosarcoma and undifferentiated pleomorphic sarcoma are aggressive cancers that tend to metastasize, threatening the lives of children and adults. These types of sarcoma may be particularly vulnerable to immunotherapies.

Targeting Undifferentiated Pleomorphic Sarcomas

by Dr. Torsten Nielsen

Undifferentiated pleomorphic sarcomas never did have any consistent, recognizable genetic changes we could target, and so were not a focus for much sarcoma research. Immunotherapy approaches in some way are admitting that we can't figure out how to target a cancer, but maybe the patient's own immune system can do that for us, if only we give it a helping hand. The very aspects of undifferentiated sarcomas that made them so hard to target makes them also a particularly good sarcoma type upon which to test immune-oncology treatments.

Dr. Neilsen explains: "Both these sarcoma types (the most common soft tissue and primary bone sarcomas) have genetically complex, mutation-filled DNA, and produce large numbers of mutant proteins. These sarcomas are therefore more likely to be recognized as foreign and consequently to attract immune responses."

The ImmunoSarc Study

The investigators aim to learn how the immune system reacts to sarcomas and how the tumors manage to evade the immune system and continue growing. Dr. Blay states that the overall goal of the research is "to understand how immune cells, white blood cells, contribute to tumor progression and can be manipulated to eradicate cancer." Based on this new knowledge, the investigators can recommend treatment strategies that should help the immune system kill the tumors.

One such strategy is to use checkpoint inhibitors, drugs that block a cell's "don't kill" signal and engage the immune system. The hope is that matching the right checkpoint inhibitor to a sarcoma subtype will enable those sarcoma patients to benefit from immunotherapy.

Dr. David Thomas says that scientists are excited about newly developed checkpoint inhibitors: "These agents 'unleash' the capacity of our own immune cells, specifically T-cells, to kill cancer cells." He will be working to discover which T-cells, among the billions that humans carry, actually kill sarcomas. This information can be used in a second immunotherapy treatment strategy: the design of vaccines that are specific to individual cancer types.

The Potential of Immunotherapy and the Importance of Collaboration

According to Dr. Thomas, the most extraordinary aspect of immunotherapies in other cancers has been the duration of the patients' responses. He says, "When patients are treated with standard chemotherapy, the cancers have a tendency to come back after treatment ends. With some patients on immunotherapy, the immune system continues to eradicate cancer in an ongoing way."

Dr. Robert Maki notes that investigators have been interested in advancing immune research on sarcomas for several years, but progress can be slow in rare diseases. This International Collaborative Grant will help to ensure that sarcoma patients are not left behind as immunotherapy science advances.

Dr. Demicco explains: "Because sarcomas are so rare, it is difficult for any one institution to have adequate case numbers for a meaningful study. This granting program encourages researchers to assemble interested and enthusiastic collaborative partners with materials and resources that may not be available at their home institutions. This sharing of ideas, materials, data, and expertise creates a synergy crucial for studies of rare diseases such as sarcoma."

The Investigators

Each investigator involved in this study has an established infrastructure of equipment, specimens, and supporting personnel for sarcoma research. In addition, each member of the research team has demonstrated a track record of productivity and accomplishment in studying sarcomas. Investigators include:

  • Torsten O. Nielsen (MD, PhD, FRCPC): a professor of pathology and laboratory medicine at the University of British Columbia, a clinician-scientist and musculoskeletal consultant pathologist based at the Vancouver Coastal Health Research Institute and the British Columbia Cancer Agency.
  • Jean-Yves Blay (MD, PhD): a professor of medical oncology at the University Claude Bernard Lyon I, a medical oncologist and current Director General of the Comprehensive Cancer Center of Lyon, the Centre Léon Bérard.
  • Elizabeth Demicco (MD, PhD): an assistant professor of pathology at the Mount Sinai Hospital in New York City.
  • Robert Maki (MD, PhD, FACP): a professor of medicine, pediatrics, and orthopaedics at Mount Sinai Hospital in New York City.
  • David Thomas (FRACP, PhD): the director of the Kinghorn Cancer Centre and head of the cancer division at the Garvan Institute of Medical Research in Sydney, Australia.
The Funding

This International Collaborative Grant was co-funded by the Liddy Shriver Sarcoma Initiative ($290,400), the Alan B. Slifka Foundation ($25,000) and the Wendy Walk ($25,000). These funds, in addition to $459,600 which the investigators are focusing on this research, means that $800,000 will be used to support these studies. We hope that this research will make important and useful contributions to the understanding of the use of immunotherapeutic approaches in the treatment of undifferentiated pleomorphic sarcoma and osteosarcoma.

ImmunoSarc: Understanding the Immune Response in Sarcomas

Introduction

In the past 40 years, there has been remarkable progress in the understanding of the immune system and ways to turn it on and off to treat diagnoses as diverse as autoimmune disease and cancer. Most recently, engineered T-cells and immune checkpoint inhibitors have demonstrated activity in melanoma, non-small-cell lung cancer, and other carcinomas. Given these exciting developments in immunology and immunotherapy, we will examine sarcomas and the existing immune responses against them in greater detail than ever before.

Immune Checkpoint Inhibitors

The immune system usually recognizes and destroys pre-cancerous cells before they ever become clinically-apparent cancer. To prevent this system from destroying normal, healthy cells (as occurs in autoimmune disease), there are a number of stops, called immune checkpoints. Through immune checkpoints, normal cells put out a “don’t kill” signal, putting the brakes on the immune response and minimizing normal tissue damage. Tumor cells can sometimes mimic immune checkpoints, preventing their usual destruction by the immune system.

Immune checkpoint inhibitors are a class of drugs that block the "don’t kill" signal, releasing the brakes on the immune system. In the absence of immune checkpoint signalling, the immune system is free to launch a full-scale attack on the abnormal cancer cells.

We hypothesize that the best anti-sarcoma immune responses with checkpoint inhibitors will be seen in sarcomas that (1) express the largest number of neoantigens and (2) express immune checkpoint markers that can be inhibited by new immunomodulatory drugs. By extension, given the near absence of mutations in translocation-associated sarcomas, we further hypothesize that aneuploid sarcomas with the greatest spontaneous mutational burden and inflammatory infiltrates will be the sarcomas with the best chance for a good clinical result from immune checkpoint inhibitors.

Inflammatory Infiltrates

Inflammatory infiltration occurs when immune cells penetrate tissue (such as a tumor) to employ their immune functions. Inflammatory infiltrates consist of a number of white blood cell categories:

Lymphocytes: white blood cells involved with controlling the specificity of the immune response by first recognizing specific features (antigens) on the infectious or abnormal cells before mounting a reaction. These include T cells, B cells, and natural killer cells.

  • Cytotoxic T cells (TC cells): equipped to recognize and kill virus-infected and cancer cells
  • Regulatory T cells (Treg cells): for suppression of TC cell activity against normal cells, to prevent autoimmune attach on “self” cells
  • Helper T cells (TH cells): function to amplify the immune response by helping B cells, TC cells, and macrophages
  • Natural killer cells (NK cells): designed to quickly kill virus-infected and cancer cells, usually without antigen specificity

Macrophages: large white blood cells that engulf and destroy infectious organisms, cancer cells, and cellular debris

Dendritic cells: tissue-dwelling immune cells that engulf infectious/abnormal cells and present their antigens, to stimulate lymphocytes

Mast cells: tissue-dwelling immune cells that release chemicals to enhance inflammation and to attract more immune cells

To test our hypothesis, we will focus especially on two aneuploid sarcomas: Undifferentiated Pleomorphic Sarcoma and Osteogenic Sarcoma (Osteosarcoma). To determine if these sarcoma subtypes are genuinely good targets for immune checkpoint inhibitors we will:

  1. Examine undifferentiated pleomorphic sarcoma and osteosarcoma and associated immune infiltrates for immune regulatory molecules
  2. Study the T cells and tumors in an osteosarcoma mouse model system for neoantigens and the T cells that recognize them
  3. Characterize the neoantigens and T cells in people with osteosarcoma and undifferentiated pleomorphic sarcoma

These studies will provide the translational science underpinning for the use of checkpoint inhibitors in the management of sarcomas. The analyses conducted here may be replicated to inform use of specific immunotherapies in other sarcoma subtypes.

Background

Despite advances in our understanding of cancer biology and the development of standardized, effective treatment with surgery and radiation, sarcomas metastasize in about half of patients. Once this happens, cures are rare, even with new generations of therapies that target the underlying molecular drivers. Next generation sequencing has revealed that cancer genomes are even more complex than expected, with many subclones within each tumor. These subclones are genetically unstable and constantly generating new mutations that are selected-for in a Darwinian fashion, leading to resistance against virtually all conventional cytotoxic, hormonal, small molecule, or antibody-based targeted therapies. Even the sarcomas that affect children and adolescents, which often start out genetically simple, can adapt in this fashion.1

This problem is ultimately unsolvable without an anti-cancer approach that evolves as rapidly as tumors do. Given the limited arsenal of existing anti-cancer drugs and given that drug development takes decades (for diseases that progress in weeks), the practical answer is to harness the body’s immune system. The generally-accepted principle has been that (1) cancers express different self-antigens, and so they are not recognized as foreign; and/or (2) cancers have developed immune escape mechanisms well before they grow to a clinically-appreciable size. Consequently, immune stimulation has long been thought to be a futile approach.

It is clear that the mutation burden of a tumor impacts the immune response against that mutated tumor. In key experiments, tumors developing in immunocompetent mice could be grown in immunodeficient mice, but tumors generated in immunodeficient mice do not grow well in immunocompetent mice of the same background strain.2 More careful analysis shows a loss of immunogenic antigens in cancers transplanted from immunodeficient to immunocompetent mice, indicating a selective pressure on the cancer: cancer immunoediting.

Cancer Immunoediting3,4

The process by which cancer cells evolve to evade the anti-cancer immune response, thought to work in three phases:

  1. Elimination: The immune system recognizes and kills abnormal cells (known as immunosurveillance).
  2. Equilibrium: Some tumor cells persist but are kept in check by the immune system. The least immunogenic cells survive best.
  3. Escape: The previous balance between tumor growth and immune response shifts toward tumor growth, either due to exhaustion/inhibition of the immune system or due to new tumor cell variants that can evade or manipulate the immune system.

Following immunoediting, a cancer can grow rapidly, unchecked by the immune system.

Given these changes in perspective, cancer immunotherapy was Science magazine’s 2013 advance of the year.5 Passive immunotherapy has proven remarkably successful in selected cancer subtypes, such as lymphoma. Strategies to reactivate anti-tumor immune responses with drugs targeting CTLA4 and PD1/PDL1 have led to striking advances in the therapy of melanoma, some carcinomas, and hematopoietic neoplasias.

Inhibitory Immune Checkpoints

Immune signalling molecules that act to shut down or dampen the immune response.

  • CTLA4: A receptor present on the surface of cytotoxic T cells that dampens the immune response by reducing T cell population growth. It is triggered by ligands CD80 or CD86 (molecules that tumor cells can learn to express).
  • PD1: A receptor present on the surface of cytotoxic T cells that triggers apoptosis (“cell suicide”) of the T cells. It is triggered by ligands PD-L1 (can be present on tumor cells) or PD-L2 (present on macrophages and dendritic cells).
  • LAG3: A receptor present on the surface of both regulatory T cells (activates their anti-immune function) and cytotoxic T cells (reduces population growth).
  • BTLA: A receptor present on T cells that dampens the immune response by reducing T cell population growth and decreasing production of cytokines (which enhance the immune response). It is triggered by ligand HVEM (on macrophages and dendritic cells).
  • TIM3: A receptor present on the surface of T cells that triggers helper T cell apoptosis, promoting immune tolerance. It is triggered by ligand galectin-9 (can be present on tumor cells).deve

Immunotherapy strategies for people with sarcomas6 are comparatively less advanced than in other cancers. Evidence from mouse models7 and the observation that immunosuppressed patients have three times the incidence of (non-Kaposi) sarcomas8 support a major role for the immune system in preventing the development and progression of these cancers. Osteogenic sarcoma represents a specific sarcoma in which immunotherapy has proven effective, such that regulatory authorities have approved muramyl tripeptide for human use in primary osteogenic sarcoma. New strategies have been developed based on immune checkpoint inhibitors and tumor antigen targeting using cancer vaccines, adoptive T-cell transfer, and chimaeric antigen receptors. Thus, there is an immediate opportunity to assess the potential for such strategies to work across the spectrum of human sarcomas.

Immune Stimulation Strategies

Immunotherapy strategies that act by initiating or enhancing the immune response toward the tumor:

  • Muramyl Tripeptide: An anti-cancer drug that stimulates enhanced immune activity (in a non-specific way) by mimicking an antigen usually found on the cell wall surface of bacteria, thus triggering a generalized immune response.
  • Cancer Vaccine Treatment: In this approach, patients are “vaccinated” with antigenic proteins from their cancer cells, in hopes of triggering an immune response that specifically targets the cancer and its antigens.
  • Adoptive T-cell Transfer: T cells are collected from a surgically-removed tumor, grown outside of the body to large numbers, and re-introduced to the patient. Because they were infiltrating the tumor, these T cells are presumed to be cancer-cell-specific, and so ideally, the patient receives a large dose of immune cells that directly target any residual cancer cells (or metastases).
  • Chimaeric Antigen Receptors: Artificial T cell receptors (that recognize tumor cell antigens) are added to a patient's T cells by genetic engineering. The modified T cells are then re-introduced to the patient, similar to adoptive T-cell transfer.

We will focus much of the work on the antigens found within sarcomas and the immune checkpoints that appear to regulate existing immune responses against neoantigens. Immune checkpoints are crucial for maintaining self-tolerance and limiting immune responses in peripheral tissues; they are also involved in allowing immune tolerance of tumors. There is growing interest in the immunoregulatory receptor PD1 and its large family of ligands as a potential mechanism of tumor immune tolerance and escape. In the inflammatory setting, stimuli such as IFNγ may upregulate PD-L1 expression in peripheral tissues and in immune cells to suppress the immune response. Many different cancers (melanoma, ovarian cancer, colorectal cancers) up-regulate PD-L1 constitutively or in response to inflammation.

Little is known to date concerning immune checkpoint mechanisms in sarcomas. So far, one study reporting the expression of PD1/PD-L1 in osteosarcoma showed that 84% of the osteosarcoma samples expressed PD-L1, among which 24% had very high levels of PD-L1 transcript. This finding suggests that PD-L1-directed therapies could be relevant for a subset of patients with osteosarcoma.

In a similar fashion, very little is known about the potential neoantigenic repertoire of aneuploid sarcomas. There are precious few undifferentiated pleomorphic sarcoma and osteosarcoma samples in The Cancer Genome Atlas (TCGA repository), and there is no data regarding T-cell receptor usage in these tumor samples.

In the ImmunoSarc program, by gathering a group of international researchers/centers with large accrual and core research structures in place, we will fill this pre-clinical knowledge gap. This new data should, at a minimum, improve clinical trial design, efficiency, and disease prioritization for the coming wave of sarcoma immunotherapy trials.

The Investigative Team

Torsten O. Nielsen (MD, PhD, FRCPC): is a Professor of Pathology and Laboratory Medicine at the University of British Columbia, a clinician-scientist and musculoskeletal consultant pathologist based at the Vancouver Coastal Health Research Institute and the British Columbia Cancer Agency. He co-chairs the Sarcoma Disease Site Committee for the NCIC-Clinical Trials Group, and serves as associate director of the MD/PhD program for his university. Dr. Nielsen has been active in sarcoma research for the past 15 years, translating genomic profiling results into new diagnostics, predictive biomarkers, experimental therapeutics and clinical trials. He also has an active research program in breast cancer, focusing on the development and validation of biomarker tests, including for immuno-oncology.

Jean-Yves Blay (MD, PhD): is a Professor of Medical Oncology at the University Claude Bernard Lyon I, Medical Oncologist, and current Director General of the Comprehensive Cancer Center of Lyon, the Centre Léon Bérard, President of the French Sarcoma Group and of the NETwork for expert centers in SARcomas, Director of the Integrated Site for Innovative Research in Oncology in Lyon (SIRIC), Past-President of the European Organization of Research and treatment of Cancer, corresponding member of  the French Academy of Medicine. His research work in the last 15 years has been focusing on basic (molecular nosology), translational (identification of novel targets), and clinical research (new drugs development and novel strategies) on sarcoma, along with translational research on the interactions of cancer cells and immune cells in breast and ovarian carcinoma models, in particular the mechanisms of immune deactivation. This work is conducted within the Unit 17 of the Cancer Research Center of Lyon, co-led with Dr. C Caux. The ImmunoSARC project merges these two aspects of the research themes.

Elizabeth Demicco (MD, PhD): is an Assistant Professor of Pathology at the Mount Sinai Hospital in New York City. Her clinical and research focus is on mesenchymal neoplasia, with particular interest in translational studies in sarcoma. Since 2010, she has authored or co-authored over 20 papers on bone and soft tissue sarcomas.

Robert Maki (MD, PhD, FACP): is a Professor of Medicine, Pediatrics, and Orthopaedics at Mount Sinai Hospital in New York, NY, USA. He has worked in sarcoma medical oncology for over 15 years, with research experience in sarcomas for over 25 years, including a PhD in immunology examining innate and adaptive immunity in sarcomas. He has interests in sarcomas of soft tissue and bone, their biology, and in clinical trials and new drug development for this diverse group of diseases. He has mentored over 25 fellows and MD/PhD students and continues to develop collaborative clinical and translational studies.

David Thomas (FRACP, PhD): is the Director of the Kinghorn Cancer Centre and Head of Cancer Division at the Garvan Institute of Medical Research in Sydney, Australia. Dr. Thomas has a particular focus on the impact of genomics on cancer medicine, and the immunobiology of sarcomas.

Aims of the Integrated ImmunoSarc Research Program

The overall objective of the ImmunoSarc program is to interpret sarcoma biology in the context of the improved understanding of the immune response in cancer. Specifically, we will identify potential antigen targets harbored by sarcomas, as well as describe and assess the meaning of immune infiltrates observed in various sarcoma subtypes, emphasizing immune checkpoint proteins and assessing the potential role of immunotherapies in these diseases.

Within this theme, our aims are:

  1. To assess and validate immune biomarkers and immune system infiltrates in specific sarcoma subtypes
  2. To evaluate the immune response in genetically engineered mouse models (GEMMs) and in humans with osteosarcoma
  3. To explore the evolution of sarcomas in relation to immunoediting and neoantigens

Figure 1: Flow of information and tissue resources between institutes...

Figure 1: Flow of information and tissue resources between institutes...

Figure 1 outlines the flow of information and materials for this international collaboration. In brief, Aim 1 will use materials from New York and Canada for biomarker screening and assay development, and materials from Canada and France for validation. This will be co-led by Demicco and Nielsen, with support from pathologists affiliated with the French group (Blay). Aim 2, co-led by Thomas and Blay, will use assay methods developed in Aim 1 for immune cell subset and checkpoint biomarkers, with pathology support from Demicco and Nielsen. Aim 3, led by Maki (with support from Thomas and Blay), will analyze French specimens using the facilities available in New York.

Aim 1. Assessment and validation of immunologic biomarkers and immune system infiltration in specific sarcoma subtypes

This will be the world’s most comprehensive survey describing immune infiltrates across sarcomas, by virtue of access to large banks of patient tumor tissues from Canada, New York, and France:

  • The Genetic Pathology Evaluation Centre at the University of British Columbia has collected a series of 1,809 mesenchymal tumors, already assembled into tissue microarrays and used for the assessment of immunohistochemistry biomarkers.9,10 The original specimens and follow-up information are available for these patients.
  • Mount Sinai Hospital has access to patient tissue from 700 mesenchymal tumors. Tissue microarrays have been assembled for undifferentiated pleomorphic sarcoma, osteosarcoma, and synovial sarcoma.
  • Conticabase (France) includes 14,205 mesenchymal tumors from patients treated at an expert center, with regular follow-up and updated clinical information. Tissue microarrays have been constructed for over 400 patients, focusing on undifferentiated pleomorphic sarcoma and leiomyosarcoma.

Using commercial antibodies, we will perform antibody staining on the sarcoma tissues, and establish a scoring system for each biomarker. The Conticabase tissue microarrays – of special value due to their large size and detailed linked clinical data – will be used to validate our findings.

Through this aim, we will, more thoroughly than ever before:

Aim 1a. Quantify the immune infiltrates, by simple H&E assessment. This is an inexpensive, practical method to associate immune response to sarcoma subtype, which can be applied in any laboratory. Pathologists can easily identify and count overall levels of tumor-infiltrating lymphocytes. While certain sarcomas are known to be associated with tumor-infiltrating lymphocytes, a systematic survey using consistent analytical methods has not been performed. Standard methods for tumor-infiltrating lymphocyte assessment will be applied, as disseminated recently by an international group, including Nielsen.12

Aim 1b. Characterize the immune infiltrates by subtypes, including lymphocytes, macrophages, dendritic cells, and mast cells. Studies in carcinoma and melanoma have shown that the composition of the immune infiltrate has biologic, prognostic, and therapeutic significance. Tumor-infiltrating lymphocytes, for example, include cytotoxic, regulatory, helper, and gamma-delta T cells, natural killer (NK) cells, and B cells that cannot be distinguished by H&E staining. These (and other immune infiltrates) are instead distinguished by the immunohistochemistry antibodies (Table 1). This work will characterize the number and type of tumor-infiltrating lymphocytes, tumor-associated macrophages, dendritic cells (the most efficient antigen-presenting cells (APCs)) and mast cells present in human sarcomas. Sarcoma types will then be ranked by total, cytotoxic, and regulatory immune cell infiltrates, as a guide for which histologies are indeed the most immunogenic. The presence and types of infiltrates will also be linked to prognosis.

Aim 1c. Describe expression of immune regulatory molecules and their ligands, on immune infiltrates and sarcoma cells. Studies in melanoma and carcinoma have identified a series of important immune cell receptors and their ligands that are critical in stimulating or dampening the immune response. New therapies that target these molecules have recently become available. The cell surface receptor and ligand proteins we are interested in, along with their existing therapies, are listed in Table 2a. Briefly, these include:

  • The tumor necrosis factor receptor (TNFR) superfamily, which activate anti-tumor immune responses.15,16
  • The B7-CD28 superfamily of immune checkpoints, which include CTLA-4, PD1 and PD-L1/2, BTLA, ICOS, and B7-H3/4.
  • Additional lymphocyte co-regulators, including LAG3, TIM3, NKG2/2D, and the KIR family receptors.
  • Cytokines and their receptors, including IL-12 and IL-23 (secreted immunostimulatory factors), IL-6 and IFNα/β (discussed in detail in Aim 2), and CSF1R (macrophage stimulating receptor).

Major histocompatibility complex (MHC) proteins expressed on sarcoma cells (principally HLA-A/B/C and -E), which can be downregulated as an immune escape mechanism in aggressive Ewing sarcoma and osteosarcoma cases.25.26

To date, studies of these biomarkers in sarcoma are limited to unvalidated small series covering only a few subtypes. We have critically reviewed the literature and identified protocols (antibody, conditions, scoring methodology) for most of these proteins, as detailed in Table 2b.

Aim 1d. Identify tumor-associated antigens and neoantigens considered to be immunogenic and targetable with new therapies. Some tumor types – such as synovial sarcoma and myxoid liposarcoma – generally lack immune infiltrates, implying that they may be inherently immunoevasive. For these tumors, the issue is not bypassing regulatory immune checkpoints, but rather, making the immune system recognize and react to the cancer in the first place. Interestingly, these tumor types do express CT antigens, including SSX and NY-ESO-1. We will survey sarcomas for tumor cell expression of these and other immunogenic, targetable antigens.

Aim 1e. Employ expression profiling as an alternative to immunohistochemistry, which only assesses single biomarkers in a semi-quantitative manner, with somewhat subjective interpretation. Gene expression profiles produce quantitative, robust immune signatures in a highly reproducible way. NanoString is a new technology that can investigate the expression level of a specified set of genes, often as panels of related genes. It can be applied to paraffin-embedded tissue (such as the tissue microarray source blocks), and fortunately, an immune expression panel has recently become available.29 Therefore, for those sarcoma subtypes that consistently demonstrate the most immune infiltrates, this technology will be applied in a pilot study of 100 cases, to more thoroughly characterize the immune infiltrate components and active pathways.

Expected Outcomes of Aim 1

This work will yield the largest comprehensive analysis of immune infiltrates and related biomarkers in sarcomas. Patients presenting with high levels of tumor-infiltrating lymphocytes, macrophages, and NK cells are thought to have "immunologically primed" tumors, which are the most likely to respond to immune checkpoint therapies. Further, tumor types lacking such responses but expressing targetable antigens represent "immunoevasive" tumors, which may be better subjected to immune stimulation strategies.

Aim 2. Evaluation of the immune response in genetically modified mouse models (GEMM) and humans with bone sarcomas.

While Aim 1 will provide a description of potential avenues for successful application of immunotherapy in sarcoma, an experimental model is needed to better understand the mechanisms by which sarcomas escape immunosurveillance and how these mechanisms may be overcome with appropriate immunotherapy. One such model has been developed for osteosarcoma by Dr. Thomas’ group.

Model System

Figure 2: To induce osteosarcoma, mice (strain C57/Bl6) were injected...

Figure 2: To induce osteosarcoma, mice (strain C57/Bl6) were injected...

As radiation is a known risk factor for the development of osteosarcoma,30 we use a radiocarcinogen-induced osteosarcoma mouse model (Figure 2). To induce osteosarcoma, mice are injected with radioactive calcium, which generates osteosarcoma in approximately 100% of mice,31,32 with latency around 8 months. These mice are immunocompetent, and are thus an ideal model in which to study the anti-tumor immune response. Moreover, the use of radiation to induce tumors mimics the complex genomes of human tumors.

Using Rb+/- mice as the genetic background for 45Ca-induced osteosarcoma, we have shown that a number of cytokines – including IL-6 – are important for maintaining RB1 tumor suppressor function. IL-6-deficient mice are predisposed to developing radiation-induced osteosarcomas, suggesting that IL-6 also functions as a tumor suppressor.35

Cytokines

Cytokines are small secreted proteins that serve as signalling factors to other cells; in this case, to help mediate the immune response.

  • IL-6: plays a role in immune cell development and in the immediate response to infection or tissue damage.37  It is secreted by many cell types – including osteoblasts37,38 (bone-building cells) – and can both inhibit39 and promote40 tumor development.
  • IL-12: a pro-inflammatory cytokine that stimulates the growth and function of T cells, as well as promoting secretion of other pro-inflammatory cytokines. It is secreted mostly by antigen-presenting cells such as dendritic cells and macrophages.
  • IL-23a: a pro-inflammatory cytokine that promotes increased blood flow to the area. It is also secreted by antigen-presenting cells.

Figure 3: Tumors grown in wild-type (WT = normal mice) and in IL-6-deficient...

Figure 3: Tumors grown in wild-type (WT = normal mice) and in IL-6-deficient...

Behavior of individual tumors was further studied using an allograft model of tumors derived from the radiation-induced osteosarcomas. Tissue from tumors grown in wild-type and in IL-6-deficient (IL-6-/-) mice were transplanted to both wild-type and IL-6-deficient mice. Of note, tumors from IL-6-deficient tumors showed reduced growth when implanted in wild-type mice (Figure 3), demonstrating that IL-6 impacts the anti-tumor immune response.

Critical to the studies proposed, the IL-6-deficient osteosarcoma allografts ultimately develop resistance to the immune suppression within wild-type hosts and begin to grow out (Figure 9). Understanding the mechanisms of this phenomenon is the primary goal of this aim.

Figure 4: Escape of IL-6-deficient (IL-6-/-) osteosarcomas grafted into wild-type mice...

Figure 4: “Escape” of IL-6-deficient osteosarcomas grafted into wild-type...

Aim 2a: Interrogate how osteosarcoma evades IL-6-mediated tumor suppression in vivo: Our preliminary studies show that the immune microenvironment puts pressure on osteosarcomas to restrict tumor growth. In response, some tumors evolve – through as yet poorly-understood mechanisms – to escape this immunosurveillance. These mechanisms may be either genetic or epigenetic. We propose to take advantage of syngeneic models to determine whether there are consistent differences between osteosarcomas that occur in the presence or in the absence of IL-6.

Analyses of the osteosarcoma immune microenvironment and immune checkpoint expression will be performed by immunohistochemistry. Building on knowledge developed in Aim 1, we will examine the subtypes of immune infiltrates in our mouse orthotopic osteosarcoma model. We will also evaluate T-cell activation, cytokine production (in particular IL-6, IL-23, and IL-12), and PD1/PD-L1 expression. These results will be compared to findings about the immune microenvironment in a rat osteosarcoma model and in human osteosarcoma samples (representing a wide range of clinical scenarios: primary vs. metastasis, diagnosis vs. post-treatment, primary vs. relapse).

We will next compare the whole genomes between primary osteosarcomas derived from wild-type and IL-6-deficient mice (8 each), to identify recurrent genotypic differences between tumors evolving in a more immune permissive environment versus those evolving under IL-6 tumor suppression. To compensate for tumor heterogeneity, a parallel study will be run, using at least 2 IL-6-deficient osteosarcoma cell lines, which will be implanted into wild-type and IL-6-deficient mice (see Figure 8). Tumors will be allowed to grow until spontaneous resistance emerges.  We hypothesize that at least some of the strategies for this “escape” from IL-6-mediated immunosurveillance will be identifiable using genome sequencing, and that some of the same genetic adaptations will be observed between tumors implanted in different mice. In addition, we will compare genome differences between parental cell lines and those clones that escape immunosurveillance. This strategy will provide method to distinguish passenger from driver mutations. Mouse whole genome sequencing will be performed to identify recurrent mutations. Specific attention will be paid to genes involved in cell-host signaling and on immune pathways.

Aim 2b: Identify how immune checkpoints are involved in osteosarcoma response to conventional cytotoxic chemotherapy and PI3K/Akt/mTOR-targeted therapy: Prior studies from Dr. Blay’s group showed that, in both human tumors and in a rat model, chondrosarcoma cells expressed significant PD-L1. In the rat model, therapeutic agents against the PI3K/Akt/mTOR pathway caused a decrease in PD-L1 expression. This was confirmed in vitro in different sarcoma cell lines, and will be further investigated in a study exploring everolimus treatment prior to definitive surgery.

PI3K/Akt/mTOR Pathway

A signalling pathway that controls the cell cycle, the process by which cells grow and divide. Usually, the cell cycle is tightly regulated, to prevent cells from growing out of control. Dysregulation of the cell cycle is common in cancer.
The PI3K/Akt/mTOR pathway is a series of protein interactions. Briefly:

  1. PI3K is stimulated at the cell surface by an external signal.
  2. PI3K modifies Akt, activating it and bringing it to the cell surface.
  3. Akt activates mTOR, a protein that alters the expression of genes involved in cell growth, replication, motility, and survival.

When this pathway is overactive, as it is in many cancers, there is enhanced cell division and repressed cell death.

We will extend these observations to osteosarcoma, using our in vivo mouse and rat models to investigate the impact of PD1/PD-L1 blockade on:
   1. Tumor progression, both local invasion and metastatic spread.
   2. Osteosarcoma response to conventional chemotherapy (doxorubicin and ifosfamide).
   3. Osteosarcoma response to targeted therapy against the PI3K/Akt/mTOR pathway.

In addition to tumor response, we will evaluate local and metastatic tumors treated by these various combination therapies for expression of PD1/PD-L1, alterations in tumor-infiltrating lymphocytes, and cytokine production.

If relevant, in view of the previous observations, osteosarcoma cells overexpressing PD-L1 will be generated by lentiviral transfection. After analyzing the effects of PD-L1 overexpression in vitro, the effect of PD-L1 transfection on osteosarcoma progression will be analyzed in vivo, as well as its impact on immune cell infiltrates and cytokine production.

Expected Outcomes of Aim 2

The studies in Aim 2 will establish how the osteosarcoma genome is sculpted by immune pressures in vivo, and they will specifically shed light on pathways affecting the fundamental mechanisms of immune escape. This is valuable for understanding the role of the immune system (cytokines, immune checkpoints) in eliminating or suppressing osteosarcoma, and how this relates to tumor growth and progression in an immunocompetent, syngeneic model. Combining immunotherapy with conventional chemotherapy may be rationally proposed for further pre-clinical experiments, with the aim of expanding this to the clinical setting for the development of powerful targeted therapy.

Aim 3. Investigation of immune evolution in the natural history of sarcomas

One of the ways that cancer cells are thought to become recognizable to the immune system as "foreign" (and therefore in need of destruction) is the creation of abnormal peptides (neoantigens) by driver or passenger mutations within the cancer.41 This is believed to happen more frequently in aneuploid tumors such as leiomyosarcoma and undifferentiated pleomorphic sarcoma, which have much greater mutational burdens than that seen in translocation-associated sarcomas.

The most important place to assess immune responses to tumor neoantigens is in the tumor itself.42 The presence of high levels of tumor-infiltrating lymphocytes in several types of sarcoma has been previously described and will be confirmed and characterized in Aim 1. Another way to better-understand the immune response to a particular tumor is to sequence the T-cell receptors (TcR) present on tumor-infiltrating lymphocytes and to identify if any are particularly common, reflecting recognition of tumor neoantigens. These clones can then be detected at a much lower level in the peripheral circulation. Information about the T-cell repertoire can be useful to guide therapy in patients treated with immune checkpoint inhibitors. In these cases, pre-treatment tumor samples alone would be enough to identify the T-cell repertoire that should be explored in the peripheral circulation.

T-cell Repertoire

T-cell repertoire: The number of different T-cell receptors (TcR) made by the immune system. This diversity reflects the ability of the body to respond to infectious or cancer antigens: the higher the diversity, the more likely that one or more T-cells can recognize that antigen.  Because T-cells are actively involved in anti-cancer immune response, those that recognize cancer neoantigens undergo clonal expansion (growth and replication of an activated T-cell clone) during tumor growth. Clonal expansion of T-cells can be used as an indicator of neoantigen expression by tumor cells, and can be monitored to assess changes in the immune response.

Structure of the TcR: The TcR is comprised of two protein chains, usually an alpha and a beta chain (some use a gamma and delta chain). Within each original T-cell, a unique TcR is created through random rearrangements of the TcR gene, creating wide diversity for antigen recognition.

Analysis of TcR repertoire: TcR diversity and clonality is assessed by sequencing the TcR-beta chain. RNA (cellular intermediate between a DNA gene and its protein product) is extracted from tumor tissue or peripheral blood and converted to cDNA (“complementary DNA,” a lab-made version of the original gene), which is amplified and sequenced to determine the number of unique clones and the frequency of each.

Here, we will assess the interplay between tumor antigenicity and T-cell infiltrates in human sarcoma samples during disease progression, focusing on the most common aneuploid sarcomas, which also tend to be the most likely to metastasize: undifferentiated pleomorphic sarcoma, leiomyosarcoma, and osteosarcoma. Tumor samples (50 paired primary sarcomas and their metastases), will be obtained from the Conticabase tumor bank.

Aim 3a. To compare immune infiltrates, antigen expression, and immune checkpoint markers between paired primary tumors and metastases, using methods developed in Aim 1. Data will be tied to clinical aspects of the disease, relapse, metastasis, and sensitivity to treatment. 

To better understand neoantigen expression in these sarcomas, we will then assess a subset of tumors (paired primary and metastasis) by whole exome sequencing. Once the mutations are determined, potential neoantigen generation will be assessed via virtual translation of the sequences, focusing in particular on missense mutations. These mutated protein sequences will be used to create theoretical peptides, which will be analyzed to identify those most likely to bind strongly to MHC-I molecules (which would enable the immune system to recognize them as “foreign”). This will allow us to determine the antigen diversity among individual sarcomas, and to generate a neoantigen catalog for each tumor class. We will also compare the mutation profiles of primary and metastatic tumors to determine if there is a difference in neoantigen presence in metastatic disease.

Whole Exome Sequencing

A technique by which the entire protein-coding region of the genome is sequenced. Not all of DNA is made up of genes, as much of the material is used for regulation of gene expression. Exome capture is a method for collecting the coding regions of DNA for sequencing, which is valuable for looking at gene mutations without having to sequence the whole genome.

Methods: First, DNA is extracted from tissue or peripheral blood specimens. As high quality DNA is required for analysis, spectrophotometry and agarose gel electrophoresis are used to verify the amount, quality, and integrity of the DNA. Next, DNA is cut up into smaller fragments, joined to linker segments, and captured by a set of probes that bind exome DNA sequences. The captured DNA fragments are then sequenced using high throughput (next-generation sequencing) technology. Analysis is performed using software that matches sequenced fragments with their known human genes.

Aim 3b. To characterize the spectrum of T-cell receptors within sarcomas, for primary tumors, metastases, and, where available, peripheral blood obtained at initial diagnosis and after disease relapse. We will use data from tumor genome sequencing to confirm anti-tumor responses to sarcoma neoantigens by tumor-infiltrating lymphocytes.

Comparisons will include:

  1. TcR repertoire in pre-treatment vs. treated tumor samples
  2. TcR repertoire in primary vs. metastatic disease
  3. TcR repertoire in tumor samples vs. peripheral blood mononuclear cells
  4. TcR repertoire in patients who have received immune checkpoint inhibitors

This data will allow us to determine the relative frequency of specific T-cell clones in the tumor and in the periphery, which may allow us to utilize TcR testing prospectively in future studies, if common antigens and TcR are identified.

Aim 3c. To understand the effects of pembrolizumab in advanced sarcomas. Pre-treatment tumor samples and post-treatment buffy coat mononuclear cells collected in the course of the SARC028 trial – an open label, multi-centre, single stage, phase II clinical trial of pembrolizumab in highly mutated sarcomas, including undifferentiated pleomorphic sarcoma and osteosarcoma — will undergo T-cell analyses, as above. Data will be tied to tumor response.  In addition, the most interesting of the validated biomarkers from Aim 1 will be assessed in sarcoma tissues from the SARC028 trial.

Our approach focuses on the role of immune recognition of antigens presented by MHC class I molecules only, and thus, it is not capable of describing the entirety of the complex immune response elicited by sarcomas. Nonetheless, we believe that the described approach will interrogate the most relevant part of the anti-tumor immune responses and yield important new insights into the immune response to sarcomas.

Expected outcomes of Aim 3

This work will help to delineate the evolution of immune cell infiltrates in both tissue and peripheral blood over the course of sarcoma progression from local to metastatic disease. We expect to identify immune infiltrates in primary tumors associated with higher (or lower) risk of progression, independent of classical predictors for relapse.  Moreover, we will describe in greater detail the differences between primary and metastatic sarcomas, focusing on osteosarcoma and undifferentiated pleomorphic sarcoma.

For specific sarcoma subtypes, we will be able to specify TcR usage and to link this to tumor neoantigens as possible pairs necessary/sufficient for an anti-sarcoma immune response. We will also determine the relative frequency of specific TcR in the tumor vs. peripheral circulation, to determine if it is realistic to use peripheral blood to assess immune responses within a tumor.

This will serve as a proof-of-principle for future studies in patients, when it becomes possible to design immune responses against patient-specific neoantigens. Members of our group include translational clinical trial oncologists, who would be perfectly positioned to act quickly on such information.

Clinical Translation

All proposed work contributes to a common objective of identifying the immune cell populations and antigens that are most important to expected responses in patients with metastatic sarcoma, treated with immune checkpoint inhibitors. The results of Aims 1-3 will define methods that can be applied to recently-opened sarcoma immuno-oncology clinical trials (such as SARC028). Our work will also identify which sarcoma types are most likely to benefit from immunomodulatory versus immunostimulatory strategies and suggest the best combinations of immunotherapy with other available treatment strategies. Team members, with executive roles in SARC, EORTC, NCIC-CTG, and the Australasian Sarcoma Study Group, will be able use the results to directly influence the design of sarcoma immuno-oncology trials.

By Torsten Nielsen, MD, PhD, FRCPC
Pathology and Laboratory Medicine
University of British Columbia in Vancouver, Canada

Jean-Yves Blay, MD, PhD
Centre Léon Bérard in Lyon, France

Elizabeth G. Demicco, MD, PhD
Icahn School of Medicine in Mount Sinai, New York

Robert Maki, MD, PhD, FACP
Icahn School of Medicine in Mount Sinai, New York
Sarcoma Alliance for Research through Collaboration (SARC)

David Thomas, FRACP, PhD
Kinghorn Cancer Center
Garvan Institute of Medical Research in Darlinghurst, Australia

References

1. Lagarde P, Przybyl J, Brulard C, Perot G, Pierron G, Delattre O, Sciot R, Wozniak A, Schoffski P, Terrier P, Neuville A, Coindre JM, Italiano A, Orbach D, Debiec-Rychter M, Chibon F. Chromosome instability accounts for reverse metastatic outcomes of pediatric and adult synovial sarcomas. J Clin Oncol 2013; 31:608-15.

2. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, Schreiber RD. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001; 410:1107-11.

3. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv Immunol 2006; 90:1-50.

4. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011; 331:1565-70.

5. Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013; 342:1432-3.

6. D'Angelo SP, Tap WD, Schwartz GK, Carvajal RD. Sarcoma immunotherapy: past approaches and future directions. Sarcoma 2014; 2014:391967.

7. Swann JB, Vesely MD, Silva A, Sharkey J, Akira S, Schreiber RD, Smyth MJ. Demonstration of inflammation-induced cancer and cancer immunoediting during primary tumorigenesis. Proc Natl Acad Sci U S A 2008; 105:652-6.

8. Penn I. Sarcomas in organ allograft recipients. Transplantation 1995; 60:1485-91.

9. Pacheco M, Nielsen TO. Histone deacetylase 1 and 2 in mesenchymal tumors. Mod Pathol 2012; 25:222-30.

10. Endo M, Su L, Nielsen TO. Activating transcription factor 2 in mesenchymal tumors. Hum Pathol 2014; 45:276-84.

11. Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, Kellokumpu-Lehtinen PL, Bono P, Kataja V, Desmedt C, Piccart MJ, Loibl S, Denkert C, Smyth MJ, Joensuu H, Sotiriou C. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol 2014; 25:1544-50.

12. Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, Perez EA, Thompson EA, Symmans WF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2014.

13. Cavnar MJ, Zeng S, Kim TS, Sorenson EC, Ocuin LM, Balachandran VP, Seifert AM, Greer JB, Popow R, Crawley MH, Cohen NA, Green BL, Rossi F, Besmer P, Antonescu CR, DeMatteo RP. KIT oncogene inhibition drives intratumoral macrophage M2 polarization. J Exp Med 2013; 210:2873-86.

14. Buddingh EP, Kuijjer ML, Duim RA, Burger H, Agelopoulos K, Myklebost O, Serra M, Mertens F, Hogendoorn PC, Lankester AC, Cleton-Jansen AM. Tumor-infiltrating macrophages are associated with metastasis suppression in high-grade osteosarcoma: a rationale for treatment with macrophage activating agents. Clin Cancer Res 2011; 17:2110-9.

15. Ye Q, Song DG, Poussin M, Yamamoto T, Best A, Li C, Coukos G, Powell DJ, Jr. CD137 accurately identifies and enriches for naturally occurring tumor-reactive T cells in tumor. Clin Cancer Res 2014; 20:44-55.

16. Ye Q, Song DG, Powell Jr DJ. Finding a needle in a haystack: Activation-induced CD137 expression accurately identifies naturally occurring tumor-reactive T cells in cancer patients. Oncoimmunology 2013; 2:e27184.

17. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711-23.

18. Kim JR, Moon YJ, Kwon KS, Bae JS, Wagle S, Kim KM, Park HS, Lee H, Moon WS, Chung MJ, Kang MJ, Jang KY. Tumor infiltrating PD1-positive lymphocytes and the expression of PD-L1 predict poor prognosis of soft tissue sarcomas. PLoS One 2013; 8:e82870.

19. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E, Savage KJ, Hernberg MM, Lebbe C, Charles J, Mihalcioiu C, Chiarion-Sileni V, Mauch C, Cognetti F, Arance A, Schmidt H, Schadendorf D, Gogas H, Lundgren-Eriksson L, Horak C, Sharkey B, Waxman IM, Atkinson V, Ascierto PA. Nivolumab in Previously Untreated Melanoma without BRAF Mutation. N Engl J Med 2014.

20. Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res 2011; 71:5445-54.

21. Shang Y, Li Z, Li H, Xia H, Lin Z. TIM-3 expression in human osteosarcoma: Correlation with the expression of epithelial-mesenchymal transition-specific biomarkers. Oncol Lett 2013; 6:490-494.

22. Pahl JH, Ruslan SE, Kwappenberg KM, van Ostaijen-Ten Dam MM, van Tol MJ, Lankester AC, Schilham MW. Antibody-dependent cell lysis by NK cells is preserved after sarcoma-induced inhibition of NK cell cytotoxicity. Cancer Immunol Immunother 2013; 62:1235-47.

23. Smyth MJ, Swann J, Cretney E, Zerafa N, Yokoyama WM, Hayakawa Y. NKG2D function protects the host from tumor initiation. J Exp Med 2005; 202:583-8.

24. Benson DM, Jr., Caligiuri MA. Killer immunoglobulin-like receptors and tumor immunity. Cancer Immunol Res 2014; 2:99-104.

25. Nada OH, Ahmed NS, Abou Gabal HH. Prognostic significance of HLA EMR8-5 immunohistochemically analyzed expression in osteosarcoma. Diagn Pathol 2014; 9:72.

26. Yabe H, Tsukahara T, Kawaguchi S, Wada T, Torigoe T, Sato N, Terai C, Aoki M, Hirose S, Morioka H. Prognostic significance of HLA class I expression in Ewing's sarcoma family of tumors. J Surg Oncol 2011; 103:380-5.

27. Lai JP, Rosenberg AZ, Miettinen MM, Lee CC. NY-ESO-1 expression in sarcomas: A diagnostic marker and immunotherapy target. Oncoimmunology 2012; 1:1409-1410.

28. Endo M, de Graaff MA, Ingram DR, Lim S, Lev DC, Briaire-de Bruijn IH, Somaiah N, Bovee JV, Lazar AJ, Nielsen TO. NY-ESO-1 (CTAG1B) expression in mesenchymal tumors. Mod Pathol 2014.

29. Sivendran S, Chang R, Pham L, Phelps RG, Harcharik ST, Hall LD, Bernardo SG, Moskalenko MM, Sivendran M, Fu Y, de Moll EH, Pan M, Moon JY, Arora S, Cohain A, DiFeo A, Ferringer TC, Tismenetsky M, Tsui CL, Friedlander PA, Parides MK, Banchereau J, Chaussabel D, Lebwohl MG, Wolchok JD, Bhardwaj N, Burakoff SJ, Oh WK, Palucka K, Merad M, Schadt EE, Saenger YM. Dissection of immune gene networks in primary melanoma tumors critical for antitumor surveillance of patients with stage II-III resectable disease. J Invest Dermatol 2014; 134:2202-11.

30. Huvos AG, Woodard HQ. Postradiation sarcomas of bone. Health Phys 1988; 55:631-6.

31. Finkel. MP, Jinkins, P. B., Biskis, B.O. Parameters of radiation dosage that influence production of osteogenic sarcomas in mice. National Cancer Institute Monograph 1964; 14:243-269.

32. Kansara M, Tsang M, Kodjabachian L, Sims NA, Trivett MK, Ehrich M, Dobrovic A, Slavin J, Choong PF, Simmons PJ, Dawid IB, Thomas DM. Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice. J Clin Invest 2009; 119:837-51.

33. Toguchida J, Ishizaki K, Sasaki MS, Nakamura Y, Ikenaga M, Kato M, Sugimot M, Kotoura Y, Yamamuro T. Preferential mutation of paternally derived RB gene as the initial event in sporadic osteosarcoma. Nature 1989; 338:156-8.

34. Deshpande A, Hinds PW. The retinoblastoma protein in osteoblast differentiation and osteosarcoma. Curr Mol Med 2006; 6:809-17.

35. Wunder JS, Czitrom AA, Kandel R, Andrulis IL. Analysis of alterations in the retinoblastoma gene and tumor grade in bone and soft-tissue sarcomas. Journal of the National Cancer Institute 1991; 83:194-200.

36. Kansara M, Leong HS, Lin DM, Popkiss S, Pang P, Garsed DW, Walkley CR, Cullinane C, Ellul J, Haynes NM, Hicks R, Kuijjer ML, Cleton-Jansen AM, Hinds PW, Smyth MJ, Thomas DM. Immune response to RB1-regulated senescence limits radiation-induced osteosarcoma formation. J Clin Invest 2013; 123:5351-60.

37. Bluethmann H, Rothe J, Schultze N, Tkachuk M, Koebel P. Establishment of the role of IL-6 and TNF receptor 1 using gene knockout mice. J Leukoc Biol 1994; 56:565-70.

38. Feyen JH, Elford P, Di Padova FE, Trechsel U. Interleukin-6 is produced by bone and modulated by parathyroid hormone. J Bone Miner Res 1989; 4:633-8.

39. Revel M, Katz A, Eisenbach L, Feldman M, Haran-Ghera N, Harroch S, Chebath J. Interleukin-6: effects on tumor models in mice and on the cellular regulation of transcription factor IRF-1. Ann N Y Acad Sci 1995; 762:342-55; discussion 355-6.

40. Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E, Samuels Y, Rosenberg SA. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013; 19:747-52.

41. Sherwood AM, Emerson RO, Scherer D, Habermann N, Buck K, Staffa J, Desmarais C, Halama N, Jaeger D, Schirmacher P, Herpel E, Kloor M, Ulrich A, Schneider M, Ulrich CM, Robins H. Tumor-infiltrating lymphocytes in colorectal tumors display a diversity of T cell receptor sequences that differ from the T cells in adjacent mucosal tissue. Cancer Immunol Immunother 2013; 62:1453-61.

  • Figure 1: Flow of information and tissue resources between institutes.
    Figure 1: Flow of information and tissue resources between institutes, representing interactions between centers in terms of material and intellectual resources (green). Biomarker analyses will be shared with all centers and evaluated in regular full meetings.
  • Figure 2: To induce osteosarcoma, mice were injected with...
    Figure 2. (A) To induce osteosarcoma, mice (strain C57/Bl6) were injected with radioactive calcium (45Ca), which generates osteosarcoma in ~100% of mice, with ~8 months; (B) Kaplan-Meier survival plot of Il-6-deficient (IL-6-/-) and wild-type (normal) mice (16-27 mice/group), showing that IL-6-/- mice do not survive as long as wild-type mice.
  • Figure 3: Tumors grown in wild-type and in IL-6-deficient mice...
    Figure 3: (A) Tumors grown in wild-type (WT = normal mice) and in IL-6-deficient (IL-6-/-) mice were transplanted to both WT and IL-6-/- mice (6 mice/group); Cells were implanted subcutaneously (just below the skin), and mice were monitored for tumor growth every 2-3 days; (B) Excised tumors, 21 days after transplantation, showing that tumors originating from IL-6-deficient mice did not grow well after transplantation to wild-type mice; (C) Tumor volume across time for each group of mice, quantitatively confirming (B); (D) Kaplan-Meier plot, depicting overall survival of each group of mice. Wild-type mice with tumors form IL-6-deficient mice (red) had the best overall survival after 30 days.
  • Figure 4: Escape of IL-6-deficient (IL-6-/-) osteosarcomas...
    Figure 4: “Escape” of IL-6-deficient (IL-6-/-) osteosarcomas grafted into wild-type mice. Beyond 40 days, the cancer cells begin to grow, as they have found a way to evade IL-6-mediated anti-tumor immunity.