Exploring Immunotherapies: Beyond Checkpoint Inhibitors

Similar documents
Immunotherapy Concept Turned Reality

Tumor Immunology: A Primer

Cancer Immunotherapy Survey

DISCLOSURES. Roche/MSD-Merck/Celgene: Research Funding

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

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

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

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

Emerging Targets in Immunotherapy

Immunotherapy Overview, Rationale, and Role in Clinical Practice

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

Lecture 17: Vaccines (Therapeutic and Prophylactic Types)

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

Releasing the Brakes on Tumor Immunity: Immune Checkpoint Blockade Strategies

Focus on Immunotherapy as a Targeted Therapy. Brad Nelson, PhD BC Cancer, Victoria, Canada FPON, Oct

CBER Regulatory Considerations for Clinical Development of Immunotherapies in Oncology

Viruses and cancer: Should we be more afraid?

Brave New World of Cancer Therapeutics (Back to the Future)

Immunotherapie: algemene principes

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

Society for Immunotherapy of Cancer (SITC)

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

IMMUNE EFFECTOR CELLS: CAR T AND OTHER CYTOTOXIC EFFECTOR CELLS OF THE IMMUNE SYSTEM

Developing Novel Immunotherapeutic Cancer Treatments for Clinical Use

Engineered Immune Cells for Cancer Therapy : Current Status and Prospects

CANCER 1.7 M 609,000 26% 15.5 M 73% JUST THE FACTS. More Than 1,100 Cancer Treatments in Clinical Testing Offer Hope to Patients

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

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

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

GSK Oncology. Axel Hoos, MD, PhD Senior Vice President, Oncology R&D. March 8, 2017

Tumor responses (patients responding/ patients treated)

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

Concept of oncolytic virotherapyclinical

IMMUNOTHERAPY FOR LUNG CANCER

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

Cancer immunity and immunotherapy. General principles

What's New in Oncodermatopathology: Immunotherapy Reactions

Immuno-Oncology Clinical Trials Update: Checkpoint Inhibitors Others (not Anti-PD-L1/PD-1) Issue 4 January 2017

Professor Mark Bower Chelsea and Westminster Hospital, London

CAR-T Cell Therapy: A Breakthrough Treatment for Fighting Cancer

Vaccine Therapy for Cancer

Immuno-Oncology. Axel Hoos, MD, PhD Senior Vice President, Oncology R&D. February 24, 2016

Immune Checkpoint Inhibitors: The New Breakout Stars in Cancer Treatment

AGRESSIVE LYMPHOMAS - FUTURE. Dr Stéphane Doucet CHUM

Immunotherapy of Prostate Cancer

Immunotherapy for the Treatment of Cancer

Is Prostate Cancer Amenable to Immunotherapy Approaches? New Frontiers in Urologic Oncology, September 12, 2015

Update on cell therapy using dendritic cells

Immunotherapy, an exciting era!!

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

The next steps for effective cancer immunotherapy and viral vaccines. Peter Selby FACP(UK)

Current Trends in Melanoma Theresa Medina, MD UCD Cutaneous Oncology

CANCER IMMUNOTHERAPY. Cancer Research Center, Dpt. of Medicine & Service of Cytometry University of Salamanca. IBSAL

Immuno-Oncology: Perspectives on Current Therapies & Future Developments

Rationale for Patient- Specific Vaccine Therapy for Non-Hodgkin Lymphoma

Immuno-Oncology Applications

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

DEVELOPMENT OF CELLULAR IMMUNOLOGY

Talimogene Laherparepvec (T-VEC) at the SCCA: an update

CANCER IMMUNOPATHOLOGY. Eryati Darwin Faculty of Medicine Andalas University

GENETICALLY ENHANCED CANCER THERAPIES

Immunotherapy: The Newest Treatment Route

Clinical Policy: Donor Lymphocyte Infusion

Bases biológicas de la moderna inmunoterapia en el tratamiento del cáncer

Two roads for oncolytic immunotherapy development

Immunotherapy on the Horizon: Adoptive Cell Therapy

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

Tumor Immunology. Tumor (latin) = swelling

(generic name: ipilimumab) Injection 50 mg ( Yervoy ), a human anti-human CTLA-4 monoclonal. August 21, 2018

The Emerging Role of Immunotherapy in Cancer Care Renato V. La Rocca, MD, FACP Norton Cancer Institute Louisville, Kentucky

NATURAL KILLER T CELLS EBOOK

THE ROLE OF TARGETED THERAPY AND IMMUNOTHERAPY IN THE TREATMENT OF ADVANCED CERVIX CANCER

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

Personalized medicine - cancer immunotherapy

Moving Forward with Immunotherapies for Cancer

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

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

Adverse effects of Immunotherapy. Asha Nayak M.D

Experimental Therapeutics I

Connections Between Autoimmune Disease & Cancer

Reprogramming Tumor Associated Dendritic Cells for Immunotherapy

IMMUNOTARGET THERAPY: ASPETTI GENERALI

San Diego October 27, 2018

Synergistic combinations of targeted immunotherapy to combat cancer

CANCER VACCINES THE NEXT WAVE IN IMMUNO-ONCOLOGY

Lower Baseline PSA Predicts Greater Benefit From Sipuleucel-T

(This article has multiple issues. Please help improve it or discuss these issues on the talk page.)

SUPPLEMENTARY INFORMATION

Current State of Immunotherapy for Metastatic Melanoma

Update on Melanoma Treatment. Tara C Mitchell, MD

Chapter 7 Conclusions

Darwinian selection and Newtonian physics wrapped up in systems biology

BIO 302: APRIL 3, 2014

Radiation Therapy as an Immunomodulator

Biological Therapies for Cancer: Questions and Answers

Immune surveillance hypothesis (Macfarlane Burnet, 1950s)

Where do these cells come from?

Prostate Cancer 2009 MDV Anti-Angiogenesis. Anti-androgen Radiotherapy Surgery Androgen Deprivation Therapy. Docetaxel/Epothilone

Historical overview of immunotherapy

Renal Cell Carcinoma: Systemic Therapy Progress and Promise

Transcription:

Exploring Immunotherapies: Beyond Checkpoint Inhibitors Authored by: Jennifer Dolan Fox, PhD VirtualScopics (Now part of BioTelemetry Research) jennifer_fox@virtualscopics.com +1 585 249 6231 Introduction Immunotherapy is an emerging class of cancer treatment, which stimulates the body s immune system to fight cancer. With the approval of three checkpoint inhibitors (ipilimumab, pembrolizumab and nivolumab) by the Food and Drug Administration (FDA) in the past five years, immunotherapies are seen as exciting treatments able to elicit durable cures. In a previous VirtualScopics whitepaper, Immunotherapy Concept Turned Reality [1], an introduction to the role of the immune system and an overview of checkpoint inhibitors was presented. However, checkpoint inhibitors are only one of the many possible approaches used to harness the antitumor effects of the immune system. This whitepaper will introduce other promising and emerging immunotherapeutic approaches to treat cancer, namely adoptive cell transfer (ACT), anticancer vaccines, and oncolytic viruses. Evading the Immune System The immune system identifies and destroys developing tumors thus serving as the primary defense mechanism against cancer [2]. Tumor cells express antigens, substances considered foreign by the body. The immune system identifies and eliminates tumor cells based on the expression of these tumor-specific antigens. Attacking tumor cells requires T-cells, specialized immune cells that move from the blood stream into the tumor. The number of tumor-infiltrating T-cells present in the tumor microenvironment is correlated with better prognosis in various cancer types [3]. Despite what may seem like a straight forward process for tumors to be recognized as abnormal and destroyed by the immune system, tumors develop and spread even when the immune system is otherwise functioning normally. As discussed previously [1, 4], tumors can elude immune surveillance through a number of evasion mechanisms. Adoptive Cell Transfer (ACT) ACT is a type of personalized immunotherapy that uses a patient s own T-cells to attack tumors. In one approach, T-cells are harvested from a patient s tumor and grown in vitro over the course of several weeks. During this period the tumor-infiltrating T-cells are stimulated with factors (e.g. interleukin-2 and anti-cd3 antibodies) that induce activation and proliferation. The T-cell population expands considerably, with a 1,000-5,000 fold increase in T-cells [5]. The patient is then infused with the highly 1

activated and tumor-specific T-cells. Prior to infusion, the patient receives chemotherapy to suppress the immune system and augment the antitumor efficacy of the ACT [6]. ACT has shown promise in metastatic melanoma, inducing objective response rates (i.e. the proportion of patients who receive a complete or partial response during treatment) near 50% [5]. Another related form of ACT uses modified T-cells, such as chimeric antigen receptor (CAR)-T cells. The procedure includes apheresis to isolate peripheral blood mononuclear cells from the patient s blood [7]. Once extracted, the T-cells are modified in vitro to express specialized proteins, termed CARs, on their surface. The T-cell is therefore genetically re-engineered to recognize a specific antigen found on the tumor. The process of modifying the T-cells is complex and requires gene transfer technologies [7]. Similar to tumor-infiltrating T-cells, the CAR modified-t cells are expanded in culture and reinfused into the patient. With guidance from the engineered receptor, the modified T-cells can preferentially recognize, target, and destroy cancer cells that harbor the CAR-specific antigen. Although genetically modified, CAR T-cells still maintain intrinsic T-cell functionality, including the ability to proliferate, localize to sites of disease, and elicit cytotoxic effects [7]. CAR T-cells have been used in clinical trials to treat both solid and hematologic tumors, with the greatest successes seen in leukemia and lymphoma [8-10]. Recently, a phase I study using CAR T- cells directed at the CD19+ antigen in patients with acute lymphoblastic demonstrated complete remission in 90% of patients [11]. Vaccines Cancer vaccines can be used as prophylactic measures to protect against cancer causing viruses. For example, vaccinations for hepatitis B and the human papillomavirus are given to reduce the risk of developing liver and cervical cancers, respectively [12]. However, vaccinations can also be used to treat cancer. Several vaccines strategies, including cell-based (tumor or immune), protein/peptidebased, and genetic-based (DNA, RNA and viral) vaccines, have been developed and evaluated [13]. For brevity this whitepaper will introduce two cell-based approaches, tumor cell vaccines and dendritic cell vaccines. Tumor cell vaccines can be prepared using patient-derived tumor cells [13]. From the tumor cells, antigens specific to the tumor are isolated and combined with an adjuvant. Adjuvants are substances that when combined with antigens enhance the body s immune response to the antigen [14]. This cocktail is then injected into the patient from which the tumor fragments were isolated and an immune response is triggered via dendritic and T-cell interactions. Another type of vaccine uses dendritic cells. Dendritic cells are specialized immune cells that engulf antigens. The antigen is processed by the dendritic cells and presented to naïve T cells, resulting in the priming and activation of the T-cells against cancer-specific antigens [4]. For vaccine creation, dendritic cells are removed from the blood and manipulated in vitro. Specifically, dendritic cells are given tumorassociated antigens and treated with adjuvants [13]. The antigen-loaded dendritic cells are then returned to the blood stream to prime and activate T-cells. 2

Dendritic cell vaccines have been tested in various indications including prostate cancer, melanoma, renal cell carcinoma, and glioma [13]. In 2010, sipuleucel-t (marketed as Provenge ) was approved by the FDA for the treatment of metastatic castrate-resistant prostate cancer [15]. The sipuleucel-t vaccine consists of unfractionated peripheral blood mononuclear cells (which include dendritic cells) that have been incubated with a fusion protein. The fusion protein links the cytokine, granulocytemacrophage colony stimulating factor (GM-CSF), to the prostate cancer antigen, prostatic acid phosphatase (PAP). In the randomized phase III trial, patients receiving the vaccine demonstrated a significantly increased survival benefit of 4.1 months compared to unvaccinated patients [16]. Sipuleucel-T is currently the only therapeutic vaccine to receive regulatory approval in the United States. Oncolytic Viruses Oncolytic viruses are a class of viruses that selectively infect and kill cancer cells without harming normal tissues. Oncolytic viruses have dual mechanisms of action to induce anti-tumor responses [12]. The first is direct lysis of the cancer cell by the virus. The second is the indirect activation of tumorspecific T-cells resulting from the cell lysis. Specifically, the antigens released from the dying cancer cells are released, engulfed by dendritic cells and presented to T-cells to induce cytotoxic effector T cells [4]. Oncolytic viruses can be grouped into two general classes. The first are those viruses that naturally favor tumor cells as their host. These include viruses such as autonomous parvoviruses, myxoma virus, Newcastle disease virus, reovirus, and Seneca valley virus [17]. The second class includes viruses that are also genetically modified for use in vaccines (e.g. measles virus and poliovirus), as well as those viruses that are reprogrammed to specifically target cancer cells (e.g. adenovirus and herpes simplex virus). In the last few years, there has been a spike in the number of viruses that have been genetically modified for use as oncolytic viruses. For example, talimogene laherparepvec (T-VEC) is a genetically altered oncolytic virus, based on a modified herpes simplex virus type 1, that has been used to treat late stage melanoma [18]. The T-VEC includes several gene modifications allowing the oncolytic virus to selectively replicate within tumors and produce GM-CSF to enhance systemic antitumor immune responses [18]. In the randomized phase III OPTiM trial, which evaluated T-VEC in late stage melanoma against GM- CSF, the durable response rate was significantly higher in the T-VEC group (16.3%) compared to the GM-CSF group (2.1%). The objective response rate was also significantly higher in T-VEC patients (26.4%) compared to GM-CSF participants (10.8%) [18]. The OPTiM study was the first phase III trial showing clinical efficacy of an oncolytic virus. In 2015, the FDA approved T-VEC, as the first-in-class oncolytic immunotherapy for treatment of advanced melanoma. 3

Summary The potential promise of immunotherapy to serve as a treatment strategy against cancer has long been postulated. However, recent successes in clinical trials using ACT, vaccines, oncolytic viruses, and check-point inhibitors, are positioning immunotherapies to become a key pillar of cancer treatment. Future research will undoubtedly identify new ways to harness the power of the immune system to fight cancer. 4

References 1. Dolan Fox, J., Immunotherapy - Concept Turned Reality. VirtualScopics White Paper, 2015: p. 1-5. 2. Hanahan, D. and Robert A. Weinberg, Hallmarks of Cancer: The Next Generation. Cell, 2011. 144(5): p. 646-674. 3. Zamarin, D. and J.D. Wolchok, Potentiation of immunomodulatory antibody therapy with oncolytic viruses for treatment of cancer. Molecular Therapy Oncolytics, 2014. 1: p. 14004. 4. Chen, Daniel S. and I. Mellman, Oncology Meets Immunology: The Cancer-Immunity Cycle. Immunity, 2013. 39(1): p. 1-10. 5. Svane, I. and V. EM., Achievement and challenges of adoptive T cell therapy with tumorinfiltrating or blood-derived lymphocytes for metastatic melanoma: what is needed to achieve standard of care? Cancer Immunol Immunother, 2014. 63(10): p. 1081-91. 6. Wrzesinski, C., et al., Increased Intensity Lymphodepletion Enhances Tumor Treatment Efficacy of Adoptively Transferred Tumor-specific T Cells. Journal of Immunotherapy, 2010. 33(1): p. 1-7. 7. Gill, S. and C.H. June, Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunological Reviews, 2015. 263(1): p. 68-89. 8. Brentjens, R.J., et al., Safety and persistence of adoptively transferred autologous CD19- targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood, 2011. 118(18): p. 4817-4828. 9. Kochenderfer, J.N., et al., B-cell depletion and remissions of malignancy along with cytokineassociated toxicity in a clinical trial of anti-cd19 chimeric-antigen-receptor transduced T cells. Blood, 2012. 119(12): p. 2709-2720. 10. Porter, D.L., et al., Chimeric Antigen Receptor Modified T Cells in Chronic Lymphoid Leukemia. New England Journal of Medicine, 2011. 365(8): p. 725-733. 11. Maude, S.L., et al., Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. New England Journal of Medicine, 2014. 371(16): p. 1507-1517. 12. Bartlett, D.L., et al., Oncolytic viruses as therapeutic cancer vaccines. Molecular Cancer, 2013. 12(1): p. 1-16. 13. Guo, C., et al., Chapter Seven - Therapeutic Cancer Vaccines: Past, Present, and Future, in Advances in Cancer Research, D.T. Kenneth and B.F. Paul, Editors. 2013, Academic Press. p. 421-475. 14. Melero, I., et al., Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol, 2014. 11(9): p. 509-524. 15. Cheever, M.A. and C.S. Higano, PROVENGE (Sipuleucel-T) in prostate cancer: the first FDAapproved therapeutic cancer vaccine. Clin Cancer Res, 2011. 17. 16. Kantoff, P.W., et al., Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer. New England Journal of Medicine, 2010. 363(5): p. 411-422. 17. Chiocca, E.A. and S.D. Rabkin, Oncolytic Viruses and Their Application to Cancer Immunotherapy. Cancer Immunology Research, 2014. 2(4): p. 295-300. 18. Andtbacka, R.H.I., et al., Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. Journal of Clinical Oncology, 2015. 33(25): p. 2780-2788. 5