CYTOKINE THERAPY: LESSONS LEARNED. Pierre L. Triozzi, MD Translational Hematology Oncology Research

Similar documents
Adverse effects of Immunotherapy. Asha Nayak M.D

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

Intron A. Intron A (interferon alfa-2b) Description

Intron A. Intron A (interferon alfa-2b) Description

Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel:

Hematopoietic Growth Factors Colony Stimulating Factors. Erythropoietin (Epoetin alfa). Granulocyte-macrophage colonystimulating factor (G-CSF).

Cytokines in Cancer Immunotherapy

Intron A. Intron A (interferon alfa-2b) Description

Immunology lecture: 14. Cytokines: Main source: Fibroblast, but actually it can be produced by other types of cells

Personalized medicine - cancer immunotherapy

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

6/7/16. Melanoma. Updates on immune checkpoint therapies. Molecularly targeted therapies. FDA approval for talimogene laherparepvec (T- VEC)

Immune Checkpoint Inhibitors: The New Breakout Stars in Cancer Treatment

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

Immunotherapy, an exciting era!!

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

Chapter 13: Cytokines

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

January 2008 IMPORTANT DRUG WARNING

Basis of Immunology and

9/22/2016. Introduction / Goals. What is Cancer? Pharmacologic Strategies to Treat Cancer. Immune System Modulation

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins,

Actimmune. Actimmune (interferon gamma-1b) Description

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

Immunotherapy Treatment Developments in Medical Oncology

Evolving Treatment Strategies in the Management of Metastatic Melanoma: Novel Therapies for Improved Patient Outcomes. Disclosures

Attribution: University of Michigan Medical School, Department of Microbiology and Immunology

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

Tumor Immunology: A Primer

(212) (347)

Immunotherapy for the Treatment of Melanoma. Marlana Orloff, MD Thomas Jefferson University Hospital

Immunosuppressants. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

DRUG NAME: Aldesleukin

CANCER IMMUNOTHERAPY Presented by John A Keech Jr DO MultiCare Regional Cancer Center

Cytokines. Luděk Šefc. Cytokines Protein regulators of cellular communication. Cytokines x hormones

Intron A Hepatitis B. Intron A (interferon alfa-2b) Description

Phase 1 Study Combining Anti-PD-L1 (MEDI4736) With BRAF (Dabrafenib) and/or MEK (Trametinib) Inhibitors in Advanced Melanoma

Risk Evaluation and Mitigation Strategy (REMS): Cytokine release syndrome and neurological toxicities

ONCOS-102 in melanoma Dr. Alexander Shoushtari. 4. ONCOS-102 in mesothelioma 5. Summary & closing

Nilotinib AEs (adverse events) in CML population:

CANCER IMMUNOPATHOLOGY. Eryati Darwin Faculty of Medicine Andalas University

Darwinian selection and Newtonian physics wrapped up in systems biology

The Immunotherapy of Oncology

Cancer Immunotherapy Future from the Past?

Development of a Novel IL-12 DNA-based Immunotherapy in Combination with Chemotherapy for Treatment of Advanced Ovarian Cancer

Immuno-Oncology Clinical Trials Update: Therapeutic Anti-Cancer Vaccines Issue 7 April 2017

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

Overcoming Toxicities Associated with Novel Checkpoint Inhibitor Immunotherapy. Tara C. Gangadhar, MD Assistant Professor of Medicine ICI Boston 2016

MabThera. SC. The wait is over. MabThera delivered in just 5 minutes. SC= subcutaneous injection

Interleukin-2 Single Agent and Combinations

Biological Therapies for Cancer: Questions and Answers

Immunotherapy versus targeted treatments in metastatic renal cell carcinoma: The return game?

Granix. Granix (tbo-filgrastim) Description

PRO L EUK I N (aldesleukin) FAC T SHE E T

Immune checkpoint inhibitors in Hodgkin and non-hodgkin Lymphoma: How do they work? Where will we use them? Stephen M. Ansell, MD, PhD Mayo Clinic

Developing Novel Immunotherapeutic Cancer Treatments for Clinical Use

4100: Cellular Therapy Essential Data Follow-Up Form

Cancer Immunotherapy: Active Immunization Approaches

Bendamustine is Effective Therapy in Patients with Rituximab-Refractory, Indolent B-Cell Non-Hodgkin Lymphoma

2/19/2018. The Immune System and Cancer. Fatal Melanoma Transferred in a Donated Kidney 16 years after Melanoma Surgery

Who is the Ideal Candidate for PEG Intron?

David N. Robinson, MD

Ipilimumab in Melanoma

Study No.: Title: Rationale: Phase: Study Period: Study Design: Centres: Indication: Treatment: Objectives: Primary Outcome/Efficacy Variable:

08/02/59. Tumor Immunotherapy. Development of Tumor Vaccines. Types of Tumor Vaccines. Immunotherapy w/ Cytokine Gene-Transfected Tumor Cells

NTD Vaccine Design Toolkit and Training Workshop Providence, RI January 05, 2011 Cytokines Leslie P. Cousens, PhD EpiVax, Inc.

Intron A Hepatitis C. Intron A (interferon alfa-2b) Description

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

Granix. Granix (tbo-filgrastim) Description

taking charge of your options For adult patients with metastatic melanoma

Clinical: Ipilimumab (MDX-010) Update and Next Steps

Study No.: Title: Rationale: Phase: Study Period: Study Design: Centres: Indication: Treatment: TAG IDAG Objectives:

Bihong Zhao, M.D, Ph.D Department of Pathology

THE CLATTERBRIDGE CANCER CENTRE NHS FOUNDATION TRUST. Systemic Anti Cancer Treatment Protocol. EDP + mitotane

PRODUCT MONOGRAPH. Ready-to use solution. 38 mg/ml gemcitabine (as gemcitabine hydrochloride) 200 mg / 5.3 ml, 1 g / 26.3 ml, and 2 g / 52.

gp100 (209-2M) peptide and High Dose Interleukin-2 in HLA-A2+ Advanced Melanoma Patients Cytokine Working Group Experience

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

TKIs ( Tyrosine Kinase Inhibitors ) Mechanism of action and toxicity in CML Patients. Moustafa Sameer Hematology Medical Advsior,Novartis oncology

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

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology

Chapter 10 (pages ): Differentiation and Functions of CD4+ Effector T Cells Prepared by Kristen Dazy, MD, Scripps Clinic Medical Group

Immune checkpoint blockade in lung cancer

PRODUCT MONOGRAPH GEMCITABINE INJECTION

Supplemental Appendix. 1. Protocol Definition of Sustained Virologic Response. A patient has a sustained virologic response if:

A. Incorrect! It s not correct. Synergism of cytokines refers to two or more cytokines acting together.

Oncologic Applications of Interleukin-2 (Aldesleukin) When Used as Monotherapy. Original Policy Date

Granix. Granix (tbo-filgrastim) Description

Immunotherapy for the Treatment of Cancer

New paradigms for treating metastatic melanoma

Tumor Microenvironment and Immune Suppression

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

Priming the Immune System to Kill Cancer and Reverse Tolerance. Dr. Diwakar Davar Assistant Professor, Melanoma and Phase I Therapeutics

Engineering an Immunity to Cancer: A New Era of Adoptive Cellular Therapy with Tisagenlecleucel (Kymriah) in Pediatric ALL

New insights into CD8+ T cell function and regulation. Pam Ohashi Princess Margaret Cancer Centre

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

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

Immunotherapy in Colorectal cancer

Mariano Provencio Servicio de Oncología Médica Hospital Universitario Puerta de Hierro. Immune checkpoint inhibition in DLBCL

Product: Talimogene Laherparepvec Clinical Study Report: Date: 31 October 2018 Page 1

Transcription:

CYTOKINE THERAPY: LESSONS LEARNED Pierre L. Triozzi, MD Translational Hematology Oncology Research

CYTOKINES Diverse group of soluble proteins produced by cells Affect either the same (autocrine) or another (paracrine) cell Interact with specific cell surface receptors

CYTOKINES Active at very low concentrations Many act as cellular survival factors Prevent apoptosis Mediate interactions between cells Regulate processes outside cells

CYTOKINES Pleiotrophy the same cytokine can act on several different cell types and can cause different responses, depending on the cell Redundancy several different cytokines acting on a cell can individually cause the same response Synergy if two (or more) cytokines are simultaneously present, the sum of the response together is greater than the sum of the individual responses Amplification cytokines induce the production of other cytokines Antagonism the presence of one cytokine inhibits the action of a different cytokine; one cytokine blocks the effect of the other

CYTOKINES vs HORMONES Act at a systemic level, participating in processes such as inflammation, septic shock, allergic reactions, and wound healing. However Act on a wider spectrum of target cells than hormones Not produced by specific cells organized in specialized glands Produced by cells distributed all over the body Sites of expression do not necessarily predict the sites of biological function.

IL-10 IL-12 TNFα TNFβ IL-1 IL-6 IL-10 IL-12 IL-15 TNFα IFNα IFNβ IFNγ IL-1 IFNα IFNβ TNFα TNFβ IL-2 IL-4 IL-6 IL-8 IL-9 IL-16 IL-17 IFNα IFNβ IFNγ IL-1 IL-2 IL-4 IL-6 IL-10 IL-13 IL-14 IFNα IFNβ IFNγ IL-4 IL-10 IL-3 IL-9 IL-10 IL-1 IL-3 IL-4 IL-8 IL-4 IL-4 IL-4 IL-4 IL-5 IL-10 IL-3 IL-4 IL-5 IL-5 IL-1 IL-8 IL-1 IL-8 TNFα TNFα IL-1 IL-3 IL-4 IL-10 IL-13 IFNα IFNβ IFNγ TNFα TNFβ TNFα IL-1 IL-3 IL-4 IL-8 IFNγ TNFα TNFβ IL-1 IL-6 IL-8 IL-10 IL-12 IL-15 IL-18 IFNα IFNγ TNFα EOSINOPHIL BASOPHIL T CELL MAST CELL B CELL NEUTROPHIL IL-4 IL-1 IL-6 IL-10 IL-12 IFNα IFNβ TNFα TNFβ MACROPHAGE

CYTOKINE Immune tolerance Anti-tumor immunity

CYTOKINE Shock Immune tolerance Anti-tumor immunity Clotting Cell death Lung injury GI tract injury Inflammation Organ Failure Death

CYTOKINE THERAPY Pharmacology Multiplicity of biologic effects Effects dependent on dose, route, and schedule PK of agent vs PK of biologic effects Blood levels do not necessarily correlate with clinical activity Recombinant proteins, differ from the natural substance All are immunogenic, impacts PK (toxicity/efficacy)

CYTOKINE THERAPY Pharmacology Multiplicity of biologic effects Effects dependent on dose, route, and schedule PK of agent vs PK of biologic effects Blood levels do not necessarily correlate with clinical activity Recombinant proteins, differ from the natural substance All are immunogenic, impacts PK (toxicity/efficacy) Considerable inter-individual variability

Cytokine IFN-α G-CSF GM-CSF IL-2 IFN-β IL-11 IFN-γ CYTOKINES FDA Approved Renal cell carcinoma, melanoma Multiple sclerosis Thrombocytopenia Indications Hairy-cell leukemia, CML, melanoma, Kaposi sarcoma, NHL, condyloma accuminata, hepatitis B and C Neutropenia, nonmyeloid malignancy Bone marrow transplantation Chronic granulomatous disease, osteopetrosis Year First Approved 1986 1991 1991 1992 1993 1997 1999

Cytokine IFN-α G-CSF GM-CSF IL-2 IFN-β IL-11 IFN-γ TNF-α CYTOKINES FDA Approved Renal cell carcinoma, melanoma Multiple sclerosis Thrombocytopenia Indications Hairy-cell leukemia, CML, melanoma, Kaposi sarcoma, NHL, condyloma accuminata, hepatitis B and C Neutropenia, nonmyeloid malignancy Bone marrow transplantation Chronic granulomatous disease, osteopetrosis Sarcoma (isolated limb perfusion) Year First Approved 1986 1991 1991 1992 1993 1997 1999 1998 (Europe)

Cytokine IFN-α G-CSF GM-CSF IL-2 IFN-β IL-11 IFN-γ TNF-α CYTOKINES FDA Approved Renal cell carcinoma, melanoma Multiple sclerosis Thrombocytopenia Indications Hairy-cell leukemia, CML, melanoma, Kaposi sarcoma, NHL, condyloma accuminata, hepatitis B and C Neutropenia, nonmyeloid malignancy Bone marrow transplantation Chronic granulomatous disease, osteopetrosis Sarcoma (isolated limb perfusion) Year First Approved 1986 1991 1991 1992 1993 1997 1999 1998 (Europe)

IFN-α Immunologic effects IL-15 pdc STROMAL CELLS NK IFNγ GrzB Prf IFNα T h17 IL-17 CD8 MHC I on target T h1 IFNγ CD80 CD86 MHC II MONOCYTE NEUTROPHIL VEGF MMP9 CXCR4 IL-12 DENDRITIC CELL MHC I

IFN-α Immunologic effects IL-15 pdc STROMAL CELLS NK IFNγ GrzB Prf IFNα T h17 IL-17 CD8 MHC I on target T h1 IFNγ CD80 CD86 MHC II MONOCYTE NEUTROPHIL VEGF MMP9 CXCR4 IL-12 DENDRITIC CELL MHC I Anti-proliferative effects Antiviral effects Anti-angiogenic effects

IFN-α (Interferon alfa-2b; Intron A ) Position 23 Arg instead of Gly of natural sequence Administered SC or IV Dosing High (>10 MIU) Intermediate (5-10 MIU) Low (<3 MIU) Schedule: either qd or tiw t½ = 4 hours

IFN-α Hematologic Malignancies Malignancy Hairy cell leukemia CML NHL (follicular) CTCL Myeloma Dose schedule 3 MIU/m 2 /d 5 MIU/m 2 /d 3-50 MIU/m 2 /d or tiw 3-50 MIU/m 2 /d or tiw 5-12 MIU/m 2 /d or tiw RR (%) (mean) 70-90 (80) 45-85 (58) 9-67 (47) 27-85 (48) 3-18 (15)

IFN-α Solid Tumors Tumor Melanoma Kaposi s sarcoma Renal cell Endocrine pancreatic Carcinoid Dose schedule 5-20 MIU/m 2 /d or tiw 3-20 MIU/m 2 /d or tiw 5-50 MIU/m 2 /d or tiw 3-5 MIU/m 2 /d or tiw 12-24 MIU/m 2 /d or tiw RR (%) (mean) 7-19 (18) 3-67 (30) 8-23 (15) 33-77 (50) 20-40 (20)

IFN-α Melanoma INDICATIONS AND USAGE Malignant Melanoma Indicated as adjuvant to surgical treatment in patients 18 years of age or older with malignant melanoma who are free of disease but at high risk for systemic recurrence, within 56 days of surgery. Proportion alive and relapse free 1 0.8 0.6 0.4 0.2 0 RFS IFN Observation 0 2 4 6 8 10 12 14 16 Proportion aliv e 1 0.8 0.6 0.4 0.2 0 OS IFN Observation 0 2 4 6 8 10 12 14 16 Years Years Kirkwood et al. J Clin Oncol 1996 20 MIU/m2 IV qd for 5 days weekly X 4 weeks (high dose induction) THEN 10 MIU/m2 tiw SC for 48 weeks (moderate dose maintenance)

IFN-α Toxicity Severity is dose- and time-related Acute: flu-like Fever, chills, myalgia, arthralgia, headaches Subsides with continued treatment Cumulative: constitutional Anorexia, depression, and fatigue Progressive and often dose-limiting Headache Malaise Fever Chills Fatigue Mylagia Low Back Pain Join Pain Nause Anorexia Confusion Depression 0 20 40 60 80 100 % Patients w ith Side Effects

Important Safety Information WARNING Alpha interferons, including INTRON A, cause or aggravate fatal or life-threatening neuropsychiatric, autoimmune, ischemic, and infectious disorders. Patients should be monitored closely with periodic clinical and laboratory evaluations. Patients with persistently severe or worsening signs or symptoms of these conditions should be withdrawn from therapy. In many but not all cases these disorders resolve after stopping INTRON A therapy. All patients receiving INTRON A therapy experienced mild-to-moderate side effects. Some patients experienced more severe side effects, including neutropenia, fatigue, myalgia, headache, fever, chills, and increased SGOT. Other frequently occurring side effects were nausea, vomiting, depression, alopecia, diarrhea, and thrombocytopenia. DEPRESSION AND SUICIDAL BEHAVIOR, INCLUDING SUICIDAL IDEATION, SUICIDAL ATTEMPTS, AND COMPLETED SUICIDES, HAVE BEEN REPORTED IN ASSOCIATION WITH TREATMENT WITH ALFA INTERFERONS, INCLUDING INTRON A THERAPY.

IFN-α Antibody Induction Antibodies to recombinant IFN-α in 67% Neutralizing antibodies in 33%. Often accompanied by a decrease in adverse effects Impact on efficacy?

CTL CD4 NK Cytolysis IL-2 Cytolysis IL-12Rβ2 IL-4Rα gp130 IL-2Rα IL-7Rα IL-12 IL-4 IL-6 TGFβ IL-2 TGFβ IL-7 Th1 Th2 Th17 Treg CELL SURVIVAL IFNγ IL-4, IL-5, IL-13 IL-17A, IL-17F, IL-22 TGFβ

IL-2 (Aldesleukin; Proleukin ) Mutation of free-cysteine to reduce aggregation Administered IV or SC Dosing High (>100 MIU) Moderate (5-20 MIU) Low (1-5 MIU) Ultra-low (<1 MIU) Schedule: qd X 5-7 days (with rest) t½ = 1.5 hours

IL-2 Dosing Standard high-dose regimen: 600,000-720,000 IU/kg q 8h over 5 days followed by 7-10 days of rest and then a repeat 5-day course. Responders are retreated following 2 months of rest Moderate- and low-dose outpatient regimens Better tolerated Efficacy?

IL-2 Dosing Standard high-dose regimen: 600,000-720,000 IU/kg q 8h over 5 days followed by 7-10 days of rest and then a repeat 5-day course. Responders are retreated following 2 months of rest Moderate- and low-dose outpatient regimens Better tolerated Efficacy? No consensus regarding the best way to deliver IL-2 from a risk-to-benefit standpoint

IL-2 Efficacy Melanoma RR (high-dose bolus) = 16% (CR 6%) Clinical Response Duration Renal Cell Carcinoma RR (high-dose bolus) = 15% (CR 7%) Clinical Response Duration http://www.proleukin.com/

IL-2 Efficacy Melanoma RR (high-dose bolus) = 16% (CR 6%) Clinical Response Duration Renal Cell Carcinoma RR (high-dose bolus) = 15% (CR 7%) Clinical Response Duration http://www.proleukin.com/

IL-2 Efficacy Melanoma RR (high-dose bolus) = 16% (CR 6%) Clinical Response Duration Renal Cell Carcinoma RR (high-dose bolus) = 15% (CR 7%) Clinical Response Duration Effect on overall survival? http://www.proleukin.com/

IL-2 Toxicity vascular permeability interstitial edema other cytokines, e.g. TNF Lymphoid infiltration into organs Organ system General Hematologic Gastrointestinal Hepatic N/V Diarrhea Toxicity Chills, fever Edema Anemia Thrombocytopenia transaminase bilirubin Incidence (%) 97 55 99 83 89 81 75 85 Renal Oliguria creatinine 81 81 Pulmonary Dyspnea ARDS 57 1 Cardiovascular Hypotension Arrhythmia MI 96 14 2 Neurologic Altered MS Seizures 82 2 Skin Rash Pruritus Exfoliative dermatitis 42 24 18

WARNINGS Therapy with PROLEUKIN (aldesleukin) for injection should be restricted to patients with normal cardiac and pulmonary functions as defined by thallium stress testing and formal pulmonary function testing. Extreme caution should be used in patients with a normal thallium stress test and a normal pulmonary function test who have a history of cardiac or pulmonary disease. PROLEUKIN therapy should be administered in a hospital setting under the supervision of a qualified physician experienced in the use of anticancer agents. An intensive care facility and specialists skilled in cardiopulmonary or intensive care medicine must be available. PROLEUKIN therapy administration has been associated with capillary leak syndrome (CLS) which is characterized by a loss of vascular tone and extravasation of plasma proteins and fluid into the extravascular space. CLS results in hypotension and reduced organ perfusion which may be severe and can result in death. CLS may be associated with cardiac arrhythmias (supraventricular and ventricular), angina, myocardial infarction, respiratory insufficiency requiring intubation, gastrointestinal bleeding or infarction, renal insufficiency, edema and mental status changes. PROLEUKIN treatment is associated with impaired neutrophil function (reduced chemotaxis) and with an increased risk of disseminated infection including sepsis and bacterial endocarditis. Consequently preexisting bacterial infections should be adequately treated prior to initiation of PROLEUKIN therapy. Patients with indwelling central lines and particularly at risk of infection with gram positive microorganisms. Antibiotic prophylaxis with oxacillian, nafcillin, ciprofloxacin or vancomycin has been associated with a reduced incidence of staphylococcal infections. PROLEUKIN therapy administration should be withheld in patients developing moderate to severe lethargy or somnolence; continued administration may results in coma.

WARNINGS Therapy with PROLEUKIN (aldesleukin) for injection should be restricted to patients with normal cardiac and pulmonary functions as defined by thallium stress testing and formal pulmonary function testing. Extreme caution should be used in patients with a normal thallium stress test and a normal pulmonary function test who have a history of cardiac or pulmonary disease. PROLEUKIN therapy should be administered in a hospital setting under the supervision of a qualified physician experienced in the use of anticancer agents. An intensive care facility and specialists skilled in cardiopulmonary or intensive care medicine must be available. PROLEUKIN therapy administration has been associated with capillary leak syndrome (CLS) which is characterized by a loss of vascular tone and extravasation of plasma proteins and fluid into the extravascular space. CLS results in hypotension and reduced organ perfusion which may be severe and can result in death. CLS may be associated with cardiac arrhythmias (supraventricular and ventricular), angina, myocardial infarction, respiratory insufficiency requiring intubation, gastrointestinal bleeding or infarction, renal insufficiency, edema and mental status changes. PROLEUKIN treatment is associated with impaired neutrophil function (reduced chemotaxis) and with an increased risk of disseminated infection including sepsis and bacterial endocarditis. Consequently preexisting bacterial infections should be adequately treated prior to initiation of PROLEUKIN therapy. Patients with indwelling central lines and particularly at risk of infection with gram positive microorganisms. Antibiotic prophylaxis with oxacillian, nafcillin, ciprofloxacin or vancomycin has been associated with a reduced incidence of staphylococcal infections. PROLEUKIN therapy administration should be withheld in patients developing moderate to severe lethargy or somnolence; continued administration may results in coma.

ALDESLEUKIN Antibody Low titers anti-aldesleukin antibodies in 66-74% Neutralizing antibody in <10% Impact on adverse effects? Impact on efficacy?

Mechanism of action? The molecular targets that influence the clinical response to cytokine therapy have not been defined The genetic or other host factors that differentiate responders and treatment failures are unknown.

CYTOKINE THERAPY Autoimmunity Vitiligo Autoimmune sequelae are more prominent in patients that respond to cytokine immunotherapy?

ADJUVANT IFN-α2b Autoimmunity and Response Patients with autoimmunity (n=52) Vitiligo Serum anti-thyroid, nuclear, DNA, and cardiolipin antibodies Patients without autoimmunity (n=148) Gogas et al. NEJM 2006

IL-2 Autoimmunity and Response Responders Non-responders P Vitiligo 28/58 (48%) 56/316 (18%) P=0.000001 Thyroiditis (abnormal TSH) 43/58 (74%) 176/314 (57%) P=0.01 Phan et al. J Clin Oncol 2001

CYTOKINES Clinical Trials IL-1β IL-4 IL-6 IL-12 TNF GM-CSF Ineffective Toxic (Did mitigate IL-2 toxicity) Minimal activity Toxic (growth factor for myeloma) Minimal activity Toxic Minimal activity Toxic Ineffective Toxic (hypotension) Inconsistent single agent activity Well tolerated

MELANOMA Adoptive Cellular Therapy Generate TIL Excise tumor Culture with IL-2 Assay specificity Select/expand (10 10 cells) Collect PBSC (G-CSF) Non-myeloablative FuCy Myeloablative FuCy TBI (2 v 12 Gy) Infuse TIL IL-2 PBSC IL-2 RR = 51% (n=35) RR 52% (2 Gy, n=25) RR 72% (12 Gy, n=25) Rosenberg et al. Science 2006; Dudley et al. JCO 2008

γ CHAIN CYTOKINES IL-2Rα IL-2Rβ IL-2 γ IL-4R IL-4 γ IL-7Rα IL-7 γ IL-9R IL-9 γ IL-15Rα IL-2Rβ IL-15 γ IL-21R IL-21 γ JAK-1 JAK-3 JAK-1 JAK-3 JAK-1 JAK-3 JAK-1 JAK-3 JAK-1 JAK-3 JAK-1 JAK-3 STAT-5 STAT-6 STAT-5 STAT-5 STAT-5 STAT-3 Interleukin Inducible Genes

γ CHAIN CYTOKINES Clinical Trials Cytokine Main Function Cancer Indications Clinical Trial IL-2 T cell generation and function, NK activation Melanoma, renal FDA approved IL-4 Th2 polarization, T proliferation, B cell activation None None IL-7 Pre-B and pre T cell development Solid tumors (HIV and HSCT) Phase II IL-12 NK activation, Th1 polarization Ovarian, melanoma, CTCL, NHL Phase I/II IL-15 Stimulates T and NK cells Melanoma, renal, solid tumors Phase I, I/II IL-21 Induction of B, T, and NK cells Melanoma, renal ovarian, solid tumors Phase II

γ CHAIN CYTOKINES Clinical Trials Cytokine Main Function Cancer Indications Clinical Trial IL-2 T cell generation and function, NK activation Melanoma, renal FDA approved IL-4 Th2 polarization, T proliferation, B cell activation None None IL-7 Pre-B and pre T cell development Solid tumors (HIV and HSCT) Phase II IL-15 Stimulates T and NK cells Melanoma, renal, solid tumors Phase I, I/II IL-21 Induction of B, T, and NK cells Melanoma, renal ovarian, solid tumors Phase II

γ CHAIN CYTOKINES Clinical Trials Cytokine Main Function Cancer Indications Clinical Trial IL-2 T cell generation and function, NK activation Melanoma, renal FDA approved IL-4 Th2 polarization, T proliferation, B cell activation None None IL-7 Pre-B and pre T cell development Solid tumors (HIV and HSCT) Phase II IL-15 Stimulates T and NK cells Melanoma, renal, solid tumors Phase I, I/II IL-21 Induction of B, T, and NK cells Melanoma, renal ovarian, solid tumors Phase II

NKT DC CD8 Cytoxicity NK Cytoxicity ADCC CD4 IL-21 Mo/Mx ADCC Th17 B Antibody γδ Treg GI Inflammation

NKT DC CD8 Cytoxicity NK Cytoxicity ADCC CD4 IL-21 Mo/Mx ADCC Th17 B Antibody γδ Treg GI Inflammation

IL-21 Phase II study of three dosing regimens Metastatic melanoma (N=40) RR = 22.5% (9/40 PR) RR not depended on IL-21 receptor expression or BRAF mutation status. OS = 12.4 months; PFS = 4.3 months Common adverse events were fatigue, rash, diarrhea, nausea, and myalgia. 6 patients had dose-limiting toxicities Petrella et al. J Clin Oncol 2012

IL-1 FAMILY IL-1α IL-1β IL-1Ra IL-18 IL-36Ra IL-36α IL-37 IL-36β IL-36γ IL-38 IL-33 Receptor/co-receptor IL-1RI/IL-1RacP IL-1RI/IL-1RacP IL-1RI IL-18Rα/IL-18Rβ IL-1Rrp2 IL-1Rrp2/IL-1 RAcp IL-18Rα, IL-18BP IL-1Rrp2/IL-1RAcP IL-1Rrp2/IL-1RAcP IL-1Rrp2 ST2/IL-1RAcP Property Proinflammatory Proinflammatory Antagonist for IL-1α, β Proinflammatory Antagonist for IL-36 Proinflammatory Anti-inflammatory Proinflammatory Proinflammatory? Antagonist Proinflammatory

IL-1 FAMILY IL-1α IL-1β IL-1Ra IL-18 IL-36Ra IL-36α IL-37 IL-36β IL-36γ IL-38 IL-33 Receptor/co-receptor IL-1RI/IL-1RacP IL-1RI/IL-1RacP IL-1RI IL-18Rα/IL-18Rβ IL-1Rrp2 IL-1Rrp2/IL-1 RAcp IL-18Rα, IL-18BP IL-1Rrp2/IL-1RAcP IL-1Rrp2/IL-1RAcP IL-1Rrp2 ST2/IL-1RAcP Property Proinflammatory Proinflammatory Antagonist for IL-1α, β Proinflammatory Antagonist for IL-36 Proinflammatory Anti-inflammatory Proinflammatory Proinflammatory? Antagonist Proinflammatory

Antigen MACROPHAGE IFNγ Th1 IL-18 IL-12 Antigen Th0 Th1 Th1 IL-2

IL-18 Phase 2, randomized dosing study Metastatic melanoma (N=64) RR: 2% (1/64 PR) TOXICITY Dose Group, mg/kg/d 0.01 n=21 0.1 n=21 1.0 n=22 Total N=64 No. of Subjects (%) Adverse Event Grade 3 Grade 4 Grade 3 Grade 4 Grade 5 Grade 3 Grade 4 Grade 5 Grade 3-5 Fatigue 2 (10) 2 (9) 4 (6) Abdominal pain 1 (5) 1 (5) 1 (5) 3 (5) Back pain 1 (5) 1 (5) 2 (3) Dyspnea 1 (5) 2 (10) 1 (5) 2 (9) 5 (8) Ascites 1 (5) 1 (5) 2 (3) Pleural effusion 1 (5) 2 (9) 3 (5) Polyarthritis 1 (5) 1 (2) Deep vein thrombosis 1 (5) 1 (2) Pulmonary embolism 1 (5) 1 (2) Cognitive disorder 1 (5) 1 (2) Lipase increased 1 (5) 1 (2) Tarhini et al. Cancer 2009

CYTOKINE THERAPY Reponses are infrequent (but durable) Toxicity is limiting Treatments are cumbersome Multiple injections/infusion Immunogenic

CYTOKINE THERAPY Improve cytokine delivery PEGylation Cytokine-antibody fusion molecules Recombinant virus delivery systems Combination therapy Cytokine + vaccine

PEGYLATION covalent attachment of polyethylene glycol (PEG) polymer chains to drug drug solubility circulating life Increased drug stability protection from proteolytic degradation immunogenicity and antigenicity dosage frequency Without diminished efficacy Potentially reduced toxicity Improve PK

CYTOKINE PEGYLATION Drug Composition Indication FDA Approval Pegineterferon alfa-2b (PegIntron; Sylatron) PEGylated interferon alpha chronic hepatitis C and hepatitis B; melanoma 2000 (2011 melanoma) Peginterferon alfa-2a (Pegasys) PEGylated interferton chronic hepatitic C and hepatitis B 2001 Pegilgrastim (Neulasta) PEGylated recombinant methionyl human granulocyte colonystimulating factor chemotherapyinduced neutropenia 2002 Pegylated Interferon alfa-2b

ADJUVANT INTERFERON Pegylated Interferon alfa-2b 6 mcg/kg/week SC for 8 doses THEN 3 mcg/kg/week SC for up to 5 yr Eggermont et al. Lancet 2008

ADJUVANT INTERFERON Pegylated Interferon alfa-2b Cytopenia Flu like symptoms Hepatotoxicity Neurotoxicity Fatigue Anorexia Depression HD IFN + + + + + + + PEG-IFNa2b + + + More favorable safety profile + Greater convenience (less frequent dosing) FDA approval adjuvant treatment for stage III melanoma.

IMMUNOCYTOKINES IgG-Cytokine Diabody-Cytokine scfv-cytokine Combine the specificity of antibody with the activity of cytokines

IMMUNOCYTOKINES Clinical Development Immunocytokine Format Antigen Indication Clinical Trial F16-IL2 Diabody Tenascin C A1 domain Breast, lung Phase Ib/II Hu14.18-IL2 IgG GD2 Melanoma, neuroblastoma Phase II L19-IL2 Diabody Fibronectin EDB Melanoma, pancreas, renal Phase IIb NHS-IL2LT IgG DNA Lung, NHL Phase I/II BI-IL12 IgG Fibronectin domain VII Melanoma Phase I/II NHS-IL12 IgG DNA/histone Solid tumors Phase I L19-TNF scfv Fibronectin EDB Melanoma Phase I/II

L19-IL2 (Darleukin) Immunocytokine composed of a diabody specific to the EDB domain of fibronectin, a tumor angiogenesis marker, and of human interleukin-2 (IL2). Delivers IL2 to the tumor site exploiting the selective expression of EDB on newly formed blood vessels. Eigentler et al. Clin Cancer Res 2011

L19-IL2 Clinical Trial Metastatic melanoma (N=32) RR: 8/29 (28%); 1 CR Median overall survival 14.1 months 12-month survival rate 61.5% Most frequent adverse events included chills, fatigue, and fever Dose related Mild or moderate in severity Eigentler et al. Clin Cancer Res 2011

T-VEC (Talimogene Laherparepvec; OncoVEX GM-CSF ) Attenuated herpes virus Expresses GM-CSF Recombinant virus delivery systems

T-VEC Does not replicate in normal cells Normal cells undamaged T-VEC GM-CSF Tumor antigen T Infects both tumor and normal cells DC DC Replicates and generates GM-CSF in tumors Tumor cells lyse and release tumor antigens, virus and GM-CSF GM-CSF recruits dendritic cells to tumor sties Dendritic cells generate antitumor T cells active systemically

Durable RR ( 6mo) ORR CR OS T-VEC Clinical Trial Unresectable melanoma Stage IIIB/C or IV Injectable cutaneous, SC, or nodal lesions T-VEC Intralesional N=295 16% 26% 11% 23.3 mo GM-CSF SC N=141 2% 6% 1% 19.0 mo P<.0001 P<.0001 P<.0001 P=.07 Andtbacka et al. ASCO 2013 (abstr) Well tolerated Mild fatigue, chills, or fever Most common serious adverse event was cellulitis (2%)

CYTOKINE THERAPY Combinations Despite yielding higher response rates, regimens that combine IFN with IL-2 (and/or chemotherapy) have not produced better survival

MELANOMA VACCINE HD IL-2 ± Immunization gp100:209-217(210m) peptide vaccine followed by HD IL-2 HD IL-2 P N 91 94 RR 16% 6%.03 PFS 2.2 mo 1.6 mo.008 OS 17.8 mo 11.1 mo.06 Schwartzentruber et al. NEJM 2011

CYTOKINE THERAPY One of the few approaches that can induce complete and durable tumor responses Single agent response rates have been low Dosing and scheduling have been empiric Limited by frequent and often severe toxicity Mechanism of responses are not known

CYTOKINE THERAPY Future Directions Improve PK/PD Combinations

CYTOKINE THERAPY PK/PD Prolong half-life PEGylation Enhanced specificity Cytokine-antibody fusion molecules Recombinant virus delivery systems

CYTOKINE Combinations Cytokines + chemotherapy/targeted therapy Cytokines + other immune-stimulating agents Vaccines Adoptive cellular therapy Cytokine + agents that target immunosuppression Checkpoint inhibitors (anti-ctla-4, anti-pd1) Myeloid-derived suppressor cells (e.g. sunitinib).

CYTOKINE Combinations Combination approaches will be challenging Attributing toxicity Attributing response. Some cytokines (e.g. IL-2) are thought to be most effective at high doses

CYTOKINE THERAPY Biomarkers PD/immune monitoring Cumbersome Reproducibility Identify factors that predict response Spare non-responders unnecessary treatment and toxicity

Final Rankings of Agents with High Potential for Use in Treating Cancer Rank Agent Agent Category 1 IL-15 T-Cell Growth Factor 2 Anti-PD1 and/or B7-H1 T-Cell Checkpoint Blockade Inhibitor 3 IL-12 Vaccine Adjuvant 4 Anti-CD40 and/or CD40L Antigen Presenting Cell Stimulator 5 IL-7 T-Cell Growth Factor 6 CpG Vaccine Adjuvant 7 1-Methyl Tryptophan Enzyme Inhibitor 8 Anti-CD137 (anti-4-1bb) T-Cell Stimulator 9 Anti-TGF-beta Signaling Inhibitor 10 Anti-IL-10 Receptor or Anti-IL-10 Suppression Inhibitor 11 Flt3L Dendritic Cell Growth Factor 12 Anti-GITR T-Cell Stimulator 13 CCL21 Adenovirus T-Cell Attracting Chemokine 14 Monophosphoryl Lipid A Vaccine Adjuvant 15 Poly I:C and/or Poly ICLC Vaccine Adjuvant 16 Anti-OX40 T-Cell Stimulator 17 Anti-B7-H4 T-Cell Checkpoint Blockade Inhibitor 18 Resiquimod and/or 852A Vaccine Adjuvant 19 LIGHT and/or LIGHT vector T-Cell Stimulator 20 Anti LAG-3 T-Cell Checkpoint Blockade Inhibitor NCI Immunotherapy Agent Workshop 2007