Immunotherapeutic barriers at the level of the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D.

Similar documents
Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D.

Understanding and overcoming immunoregulatory barriers within

Immunotherapeutic barriers at the level of the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D.

Molecular mechanisms of the T cellinflamed tumor microenvironment: Implications for cancer immunotherapy

Melanoma gene expression profiles to identify mechanisms of tumor resistance

Darwinian selection and Newtonian physics wrapped up in systems biology

CELLULAR AND MOLECULAR REQUIREMENTS FOR REJECTION OF B16 MELANOMA IN THE SETTING OF REGULATORY T CELL DEPLETION AND HOMEOSTATIC PROLIFERATION

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

Tumor Microenvironment and Immune Suppression

Radiation Therapy as an Immunomodulator

Disclosure Information. Mary L. Disis

Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

The development of T cells in the thymus

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

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline

VISTA, a novel immune checkpoint protein ligand that suppresses anti-tumor tumor T cell responses. Li Wang. Dartmouth Medical School

Manipulating the Tumor Environment

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center

Adaptive (acquired) immunity. Professor Peter Delves University College London

Improving cancer immunotherapy by targeting tumorinduced immune suppression

Cytokines: Interferons, Interleukins and Beyond. Michael B. Atkins, MD Georgetown-Lombardi Comprehensive Cancer Center

Advances in Cancer Immunotherapy

The Immune System. Innate. Adaptive. - skin, mucosal barriers - complement - neutrophils, NK cells, mast cells, basophils, eosinophils

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

MECHANISMS OF CELLULAR REJECTION IN ORGAN TRANSPLANTATION AN OVERVIEW

5/1/13. The proportion of thymus that produces T cells decreases with age. The cellular organization of the thymus

Developing Novel Immunotherapeutic Cancer Treatments for Clinical Use

Biologic Basis of Immunotherapy in Lung Cancer

Immune Tolerance. Kyeong Cheon Jung. Department of Pathology Seoul National University College of Medicine

Innate and adaptive immune cells in the tumor microenvironment

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Synergistic effects of indoleamine 2,3-dioxygenase (IDO) inhibitor drugs and chemotherapy in melanoma.

Cancer immunity and immunotherapy. General principles

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

Adaptive immune responses: T cell-mediated immunity

Effector T Cells and

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

SUPPLEMENTARY INFORMATION

TITLE: MODULATION OF T CELL TOLERANCE IN A MURINE MODEL FOR IMMUNOTHERAPY OF PROSTATIC ADENOCARCINOMA

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

Immunological Tolerance

5% of patients with genetic immunodeficiency develop a cancer during their lifetime (200x)

REPROGRAMING IMMUNITY IN RENAL CELL CARCINOMA

Tumors arise from accumulated genetic mutations. Tumor Immunology (Cancer)

2/16/2018. The Immune System and Cancer. Fatal Melanoma Transferred in a Donated Kidney 16 years after Melanoma Surgery

BASIC MECHANISM OF TUMOR IMMUNE SUPPRESSION

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY

Innate immune regulation of T-helper (Th) cell homeostasis in the intestine

Immunology Lecture 4. Clinical Relevance of the Immune System

Immunotherapy: The Newest Treatment Route

Tim-3 as a target for tumor immunotherapy

DNA vaccine, peripheral T-cell tolerance modulation 185

Restoring Immune Function of Tumor-Specific CD4 + T Cells during Recurrence of Melanoma

Understanding Checkpoint Inhibitors: Approved Agents, Drugs in Development and Combination Strategies. Michael A. Curran, Ph.D.

Tumor Associated Macrophages as a Novel Target for Cancer Therapy

Releasing the Brakes on Tumor Immunity: Immune Checkpoint Blockade Strategies

Immunotherapy for Malignant Brain Cancer: Overcoming Immune Suppression to Improve the Clinical Experience

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

TCR, MHC and coreceptors

Immune Checkpoint Inhibitors Drug Combinations: Patients Relevance & Ways Forward

Combining ADCs with Immuno-Oncology Agents

Bases for Immunotherapy in Multiple Myeloma

Lecture outline. Immunological tolerance and immune regulation. Central and peripheral tolerance. Inhibitory receptors of T cells. Regulatory T cells

Immunology and Immunotherapy 101 for the Non-Immunologist

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

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

Tumor Immunology: A Primer

Immune Checkpoint Inhibitors: The New Breakout Stars in Cancer Treatment

Immunotherapy, an exciting era!!

Endogenous and Exogenous Vaccination in the Context of Immunologic Checkpoint Blockade

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

Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases. Abul K. Abbas UCSF

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

Innate Immunity and the Paradoxes of Viral Pathogens and Tissue Injury in Gene Therapy

Understanding the T cell response to tumors using transnuclear mouse models

Chapter 7 Conclusions

Structured Immuno-Oncology Combination Strategies To Maximize Efficacy

Immunotherapy for the Treatment of Cancer

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

Immunotherapy in lung cancer. Saurabh maji

SEVENTH EDITION CHAPTER

Emerging Tissue and Serum Markers

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

The isbtc/sitc primer on tumor immunology and biological therapy of cancer: a summary of the 2010 program. Balwit et al.

Immunity and Cancer. Doriana Fruci. Lab di Immuno-Oncologia

Cytokine Arrays Reveal Black Ops Tactics of Tumor-induced Immunosuppression

Tolerance 2. Regulatory T cells; why tolerance fails. Abul K. Abbas UCSF. FOCiS

T Cell Activation, Costimulation and Regulation

Central tolerance. Mechanisms of Immune Tolerance. Regulation of the T cell response

Mechanisms of Immune Tolerance

Immune Regulation and Tolerance

Allergy and Immunology Review Corner: Chapter 19 of Immunology IV: Clinical Applications in Health and Disease, by Joseph A. Bellanti, MD.

Belatacept: An Opportunity to Personalize Immunosuppression? Andrew Adams MD/PhD Emory Transplant Center

Synergistic combinations of targeted immunotherapy to combat cancer

Posters and Presentations

Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells

Dendritic cells in cancer immunotherapy Aimin Jiang

Transcription:

Immunotherapeutic barriers at the level of the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D. Professor, Departments of Pathology and Medicine Program Leader, Immunology and Cancer Program of the University of Chicago Comprehensive Center

Disclosure Information Thomas F. Gajewski, M.D., Ph.D. Honoraria: BMS GSK-Bio Eisai Roche/Genentech Clinical trial grant support: BMS GSK-Bio Eisai Incyte Roche/Genentech

CD8 + cytotoxic T lymphocyte killing an antigen-expressing tumor cell

In vivo, a tumor is more than tumor cells Three dimensional mass Extracellular matrix Supported by the neovasculature, fibroblasts, macrophages Variable presence of inflammatory cells T cells (and subsets thereof) B cells/plasma cells NK/NKT cells Dendritic cell subsets The functional phenotypes of these cells may or may not be permissive for an effective anti-tumor immune response (either priming phase or effector phase) Also, likely need for dynamic interaction with draining lymph node compartment for optimal anti-tumor immunity added complexity

Model for spontaneous CD8 + T cell-mediated anti-tumor immune response in vivo MHC I MHC II Migration from tumor Immature DC granzymes B7 Tumor TCR ecd8 APC Mature DC Lymph Node ecd8 Migration to lymph node IL-2 ncd8 CD28 Migration from lymph node

Model for CD8 + T cell-mediated anti-tumor immune response in vivo: Interventions MHC I MHC II TLR ligands Migration from tumor Immature DC Blockade of suppression Vaccines B7 Costimulation Tumor TCR granzymes ecd8 Mature DC Cytokines Adoptive T cell therapy APC Lymph Node Chemokines ecd8 Migration to lymph node IL-2 ncd8 CD28 Migration from lymph node

Features of subsets of solid tumors that might mediate poor immune recognition or lack of immune destruction Priming phase Lack of innate immune-activating danger signals Poor recruitment of the critical APC subsets for cross-presentation of antigens to T cells Inadequate expression of costimulatory ligands on tumor cells or on infiltrating APCs Effector phase Inadequate recruitment of activated effector T cells Endothelial cells/homing receptors Chemokines Presence of dominant immune inhibitory mechanisms that suppress T cell effector functions Inhibitory receptors (e.g. PD-L1/PD-1) Extrinsic suppressive cells (e.g. Tregs, MDSCs) Metabolic inhibitors (e.g. IDO, arginase) Inhibitory cytokines (e.g. IL-10, TGF- )

Hypothesis Features of the tumor microenvironment could dominate at the effector phase of the anti-tumor T cell response and limit efficacy of current immunotherapies T cell trafficking into tumor Immune suppressive mechanisms at tumor site Tumor cell biology and susceptibility to immune-mediated killing Complexities of the tumor stroma (vasculature, fibrosis) Reasoned that these features could be interrogated through pre-treatment gene expression profiling of tumor site in each individual patient Such an analysis could identify a predictive biomarker profile associated with clinical response, and also highlight new biologic barriers that need to be overcome to optimize therapeutic efficacy of vaccines and other immunotherapies

Expression of a subset of chemokine genes is associated with presence of CD8 transcripts CD8 CCL2 CCL4 CCL5 CXCL9 CXCL10 CCL19 CCL21 Harlin et al. Can. Res. 2009

-3 0-2 6-2 1-1 7-1 3-0 9-0 4 0 04 09 13 17 21 26 30 Gene expression pattern of tumor microenvironment associated with favorable clinical outcome to a dendritic cell vaccine 100 Survival based on clinical response Survival groups Short Long 7 8 8 8 7 8 7 7 6 7 8 6 8 6 6 6 7 8 Survival probability (%) 80 60 40 20 0 0 10 20 30 40 50 60 70 80 Months Prolonged SD Progressive Dz No correlation with immune response in blood Thy1 CD28 CCL19 LTbeta IL-27Ralpha IL-1R T cell markers and chemokines Schuler collaboration, ASCO 2009

Inflamed gene expression signature is associated with survival following GSK MAGE3 protein vaccine 100 90 % Patients 80 70 60 50 40 30 AS15 GS+ AS02 B GS+ 20 10 AS15: HR (GS+ vs GS-) = 0.268 (95%CI [0.08;0.90]) AS02 B : HR (GS+ vs GS-) = 0.433 (95%CI [0.17;1.14]) AS15 GS- AS02 B GS- Time (months) Louahed et al., EORTC-NCI-AACR 2009

Ipilimumab clinical responders also appear to show an inflamed tumor gene expression profile CXCL9, 10, 11 CCL4, CCL5 Granzyme B Perforin CD8 No-benefit Benefit Ji et al, AACR 2011

Implication of melanoma gene array results for patient-specific therapy Gene expression profiling of the melanoma tumor microenvironment has revealed reproducible patterns associated with clinical benefit should be explored as predictive biomarker in prospective trials Already being pursued by GSK-Bio in context of multicenter MAGE3 vaccine studies Ideally, this strategy should allow enrichment for the potentially responsive patient population in the future Think Her2 equivalent for immunotherpies These observations also highlight critical aspects of tumor/immune system biology, and suggest specific strategies for overcoming immunologic barriers at the level of the tumor microenvironment

Two broad categories of tumor microenvironments defined by gene expression profiling and confirmatory assays CD8 IHC A B Gajewski, Brichard; Cancer J. 2010 T cell poor Lack chemokines for recruitment Low indicators of inflammation T cell rich Chemokines for T cell recruitment CD8 + T cells in tumor microenvironment Broad inflammatory signature Apparently predictive of clinical benefit to vaccines

1. Chemokines and T cell migration What is attracting T cells into some tumors? Can we mimic this in the tumors that fail to achieve it spontaneously?

A subset of melanoma cell lines expresses a broad array of chemokines SKMel23 M888 M537 3 4 1 2 1 2 Gro- IL-8 10 11 7 8 9 5 6 CCL2 CCL4 CCL5 CXCL9 CXCL10 Implies that in some cases, the melanoma tumor cells themselves can produce the entire panel of key chemokines

Superior recruitment of human CD8 + effector T cells in NOD/scid mice bearing chemokine-high M537 melanomas Blood Spleen Tumor ( Chemokine-high ) ( Chemokine-low )

Candidate strategies to promote effector T cell migration into tumor sites Introduce chemokines directly CXCR3-binding chemokines (CXCL9, CXCL10) Others (CCL2, CCL3, CCL4, CCL5) Induce chemokine production from stromal cells LIGHT, lymphotoxin: bind LT R Elicit appropriate local inflammation that includes chemokine production Type I IFNs TLR agonists Radiation? Alter signaling pathways in melanoma cells themselves to enable chemokine gene expression by tumor cells

Intratumoral LIGHT adenovirus in B16 melanoma: Promotes chemokine production, CD8 + T cell recruitment, primary tumor control, and rejection of non-injected distant metastases Tumor rejection CD8 + T cell infiltrate 4000 1 st tumor 2 nd tumor Tumor volume (mm 3 ) 3000 2000 1000 0 Ad-control Ad-LIGHT 0 10 20 30 10 20 30 Days after tumor challenge Ad-LIGHT Ad-control 17.2% 14.9% 4.56% 3.52% CD8 Yu et al, J. Immunol. 2007

2. T cell suppressive mechanisms Why are TIL not eliminating the tumor cells they are infiltrating? Can we overcome this defect and restore tumor rejection?

Inflamed melanomas containing CD8 + T cells have highest expression of immune inhibitory pathways IDO (indoleamine-2,3- dioxygenase) Tryptophan depletion PD-L1 Engages PD-1 on T cells CD4 + CD25 + FoxP3 + Tregs Extrinsic suppression T cell anergy (B7-poor) T cell intrinsic TCR signaling defect Immunol. Rev. 2006, Clin. Can. Res. 2007

Correlated expression of IDO, FoxP3, and PD-L1 transcripts in individual tumors

Interfering with PD-L1/PD-1 interactions can lead to tumor rejection in vivo Blank et al, Cancer Research, 2004

1-methyltryptophan reverses immunosuppression by IDO and enables tumor control in vivo Uyttenhove et al Nature Med. 9:1269, 2003

Uncoupling multiple immune suppressive mechanisms in combination: Treg depletion and anergy reversal synergize to promote rejection of B16 melanoma and vitiligo Kline et al., Clin. Can. Res. 2008

Strategies to block immune inhibitory mechanisms tested in mouse models and being translated to the clinic Blockade of PD-L1/PD-1 interactions Anti-PD-1 and anti-pd-l1 mabs (Medarex/BMS; Curetech) Depletion of CD4 + CD25 + FoxP3 + Tregs Denileukin diftitox (IL-2/DT fusion) Daclizumab or Basiliximab (anti-il-2r mab) Ex vivo bead depletion of CD25 + cells from T cell product for adoptive transfer IDO inhibition 1-methyltryptophan (RAID program) New more potent IDO inhibitors (Incyte) Anergy reversal Introduction of B7-1 into tumor sites Homeostatic cytokine-driven proliferation T cell adoptive transfer into lymphopenic recipient Exogenous IL-7 / IL-15 Decipher molecular mechanism and develop small molecule inhibitors to restore T cell function Combinations of negative regulatory pathway blockade Synergy between blockade of 2 or more pathways

Anti-PD-1 mab phase I (MDX-1106; BMS 936558): Tumor response 60.00 40.00 20.00 0.00-20.0-40.0-60.0-80.0-100 Melanoma New lesions after cycle 1 Patients Responses also seen in NSCLC and renal cell carcinoma Sznol et al, ASCO 2010

Reduction of Treg number using Denileukin diftitox can have clinical activity in melanoma Rasku et al J. Trans. Med. 2008 Multicenter phase II study currently ongoing

3. Innate immune signals type I IFNs How are anti-tumor T cells sometimes becoming spontaneously primed? Can we improve endogenous T cell priming in the tumors that fail to do so alone?

Melanoma metastases that contain T cell transcripts also contain transcripts known to be induced by type I IFNs A: IRF1 B: IFN-induced p30

Host IFN- R is critical for generating a spontaneous tumor-specific T cell response A: IFN- Rko B: Stat1ko

Mice deficient in IFN signaling fail to accumulate CD8 + DC subset in tumor microenvironment A: Percentage B: Absolute number Fuertes et al; J. Exp. Med. 2011

Anti-tumor immune responses: Working model for innate immune signals and spontaneous cross-presentation IFN- Others? CD8 DC pdc? Lymphatic CD8 DC pdc? Tumor-derived factors: HMGB1? DNA? RNA? UA? ATP? ncd8 ecd8 IL-2 Blood ecd8 ecd8 Lymph node Tumor microenvironment

Provision of exogenous IFN- can potently induce tumor rejection

IFN- signaling on host non-hematopoietic cells is necessary for control of B16 melanoma Implies effect at the level of non-hematopoietic stromal cells

Intratumoral IFN- exerts a profound anti-angiogenic effect Dark field Tumor cells (red) Endothelial cells (green) Control IFN- Spaapen et al. Manuscript in preparation

Targeting tumor stroma immunologically may be the key to durable complete responses BM chimera with MHC matched tumor, hematopoietic stroma, and non-hematopoietic stroma Spiotto, Schreiber et al. Nature Medicine 10:294, 2004

Conclusions There is heterogeneity in patient outcome to immune-based therapies for cancer such as melanoma vaccines, IL-2, and anti- CTLA-4 mab One component of that heterogeneity is derived from differences at the level of the tumor microenvironment Key determining factors in melanoma microenvironment include chemokine-mediated recruitment of effector CD8 + T cells, local immune suppressive mechanisms, and innate immune activation including type I IFNs Understanding these aspects is enabling improved patient selection for Rx with immunotherapies (predictive biomarker), and also development of new interventions to modify the microenvironment to better support T cell-mediated rejection Targeting the tumor stroma immunologically may be just as critical as targeting the tumor cells

Acknowledgments Melanoma gene array/ chemokines Helena Harlin Ruth Meng Amy Peterson Mark McKee Craig Slingluff Functional genomics core LIGHT adenovirus Yang-Xin Fu Ping Yu Hans Schreiber Uncoupling negative regulation Justin Kline Robbert Spaapen Yuan-yuan Zha Christian Blank Amy Peterson Ian Brown Type I IFNs Mercedes Fuertes Robbert Spaapen Aalok Kacha Justin Kline David Kranz Hans Schreiber Ken Murphy GSI project Ruth Meng Yuan-yuan Zha Kim Margolin SWOG, CTEP Collaborative vaccine/gene array data Gerold Schuler (Erlangen group) Vincent Brichard (GSK-Bio)

Translational research is like scuba diving Hawaii, 2011

Human CD8 + effector T cells can migrate to each of these 6 chemokines in vitro

Mechanisms of negative regulation of T cell function within the melanoma tumor microenvironment 1. Indoleamine-2,3-dioxygenase (IDO tryptophan catabolism) inhibits T cell function 2. PD-L1 (inhibitory ligand expressed by tumor cells) engages PD-1 on T cells 3. CD4 + CD25 + FoxP3 + Tregs (extrinsic suppresion) inhibit activation of effector T cells 4. T cell anergy (deficient B7 costimulation) T cell intrinsic dysfunction

IHC for IDO, FoxP3, and PD-L1 shows expression in distinct cell subsets in melanoma metastases A: IDO B: FoxP3 C: PD-L1

Implantation of tumors in vivo results in IFN- production in the tumor-draining lymph node A: IFN- mrna in DLN cells B: IFN- mrna based on CD11c