Journal of Oral & Dental Health

Similar documents
Automated manufacture of T-cell immunotherapies using gamma retroviral transduction. Lee Markwick, PhD. AMC, Manchester March 2018

Immunotherapy on the Horizon: Adoptive Cell Therapy

Sleeping Beauty: Current applications and future strategies. CAR-TCR Summit 2017 Partow Kebriaei, MD

CAR T-CELLS: ENGINEERING IMMUNE CELLS TO TREAT CANCER. Roman GALETTO, PhD 17 th Club Phase 1 Annual Meeting April 5 th Paris

Exploring Immunotherapies: Beyond Checkpoint Inhibitors

Melanoma Bridge Meeting

Abstract #163 Michael Kalos, PhD

Children s Hospital of Philadelphia Annual Progress Report: 2011 Formula Grant

Tumor responses (patients responding/ patients treated)

Advances in Adoptive Cellular Therapy of Cancer. Melanoma Bridge Meeting December 5, 2014

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

Engineering T Cells. Mack Cash

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

Chapter 7 Conclusions

NATURAL KILLER T CELLS EBOOK

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

The Adaptive Immune Responses

CBER Regulatory Considerations for Clinical Development of Immunotherapies in Oncology

Objectives. Emily Whitehead 10/11/2018. Chimeric Antigen Recepetor T-Cells (CAR-T) CAR-T Therapy: The Past, The Present, and The Future

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

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

CAR-T Therapy: The Past, The Present, and The Future. Nilay Shah, MD Michael Chargualaf, PharmD, BCOP WVU Medicine Mary Babb Randolph Cancer Center

Adoptive Cellular Therapy SITC Primer October 2012

Chimeric Antigen Receptors (CAR)

RXi Pharmaceuticals. Immuno-Oncology World Frontiers Conference. January 23, 2018 NASDAQ: RXII. Property of RXi Pharmaceuticals

Personalized medicine - cancer immunotherapy

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

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

Immunology Lecture 4. Clinical Relevance of the Immune System

Understanding the role of ex vivo T cell depletion

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

Immunocore and MedImmune announce new collaboration to conduct immuno-oncology combination trials in melanoma

Boost your immunotherapeutic research

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

Primer on Adoptive T cell Therapy. Saar Gill, MD, PhD University of Pennsylvania

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

ISCT Workshop #7 Perspectives in Cell Selection Immunomagnetic Selection

THERAPEUTIC IMPLICATIONS OF PREPARING AND ADMINISTERING INNATE IMMUNE CELLS. 9:40 am to 10:10 pm Laurence Cooper

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

National Institute for Health and Care Excellence. Single Technology Appraisal (STA)

State of the art: CAR-T cell therapy in lymphoma

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

Precision CAR T Cell Therapeutics. Carl June October 15, 2018

Artificial Antigen Presenting Cells as a Standardized Platform for Tumor Infiltrating Lymphocyte (TIL) expansion

Engineering Artificial Antigen Presenting Cells for Efficient Expansion of T Cell Subsets

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

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

GENETICALLY ENHANCED CANCER THERAPIES

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

Start your T cell research right

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

Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma

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

Cancer Immunotherapy Survey

Hosts. New Treatments for Cancer

Engineered TCR and CAR Immunotherapeutics 2015:

Simple automated manufacturing of gene engineered T cells from lymphoma and melanoma blood samples

March Corporate Presentation

Nobel Prize in Physiology or Medicine 2018

BIT 120. Copy of Cancer/HIV Lecture

Colorectal Cancer Treatment

CAR-T CELLS: NEW HOPE FOR CANCER PATIENTS

CARs vs. BiTE in ALL. David L Porter, MD Jodi Fisher Horowitz Professor University of Pennsylvania Health System Abramson Cancer Center

Cancer immunity and immunotherapy. General principles

Developing Novel Immunotherapeutic Cancer Treatments for Clinical Use

Lecture 17: Vaccines (Therapeutic and Prophylactic Types)

T-cells fighting B-cell lympho proliferative malignancies: the emerging field of CD19 CAR T-cell therapy

Connections Between Autoimmune Disease & Cancer

Advances in Immunotherapy for Lymphoma. Ashley Freeman, MD, FRCPC Immunotherapy Research Fellow

Synergistic combinations of targeted immunotherapy to combat cancer

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

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

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

Transforming patients lives through cellular immunotherapy. Next Generation Cellular Immunotherapy June 2017

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

C. Incorrect! MHC class I molecules are not involved in the process of bridging in ADCC.

Healthy cells in = good data out

Brain Tumor Treatment

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

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

Bioassays for Quality Control of Cell & Gene Therapy Products

Brian Brewer Cancer Research Institute WELCOME

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

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

Cancer Vaccines and Cytokines. Elizabeth A. Mittendorf, MD, PhD Assistant Professor Department of Surgical Oncology

LAMPvax DNA Vaccines as Immunotherapy for Cancer - Three Case Studies

Supplementary Data. Treg phenotype

Cancer Immunotherapy. What is it? Immunotherapy Can Work! 4/15/09. Can the immune system be harnessed to fight cancer? T CD4 T CD28.

xcelligence Real-Time Cell Analyzers

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

Immunotherapy of Prostate Cancer

Tumor Immunology: A Primer

Lecture 4. T lymphocytes

Emerging Targets in Immunotherapy

June IMMUNE DESIGN The in vivo generation of cytotoxic CD8 T cells (CTLs)

The Emergence of Universal Immune Receptors for Highly Personalized T cell Therapy

Cover Page. The handle holds various files of this Leiden University dissertation.

University of Lausanne and Ludwig Institute

Treatments and Current Research in Leukemia. Richard A. Larson, MD University of Chicago

The KYMRIAH Experience

Transcription:

Research Article Efficacy of Car T-Cell Therapy in Head and Neck Cancers- A Meta-Analysis Avisesh Manohar 1 *, Athira PM 1, Gaurav 2 and Shibu 3 Journal of Oral & Dental Health ISSN: 2573-8224 1 House Surgeons, NSVK Sri Venkateshwara Dental College and Hospital, Bangalore, India 2 Assistant Professor, Department of Oral Medicine and Maxillofacial Radiology, Consultant Oral Physician and Maxillofacial Radiologist, NSVK Sri Venkateshwara Dental College and Hospital, Bangalore, India 3 Reader, Department of Oral Medicine and Maxillofacial Radiology, NSVK Sri Venkateshwara Dental College and Hospital, Bangalore, India * Corresponding author Avisesh Manohar, House Surgeon, NSVK Sri Venkateshwara Dental College and Hospital, Bangalore, India, E-mail: avimanohar1996@gmail.com Submitted: 01 Feb 2019 Accepted: 22 Feb 2019; Published: 01 Mar 2019 Abstract Background: Cancer, defined by the World Health Organization (WHO) is a large group of diseases characterized by the growth of abnormal cells beyond their usual boundaries that can then invade adjoining parts of the body and/or spread to other organs. From extensive surgical excisions, radiotherapy, laser therapy to immunotherapies, various treatment strategies have been proposed and implemented so far but unfortunately none could improve the five year survival rate of the patients globally. Immunotherapy, being one amongst them, is a type of cancer treatment that boosts the body s natural defenses to fight against cancer. The current concept of immunotherapy involves Chimeric antigen receptor or the CAR T-Cell therapy which involves alterations and modifications of T cells to fight cancer cells better. Until recently, the use of CAR T-cell therapy has been restricted to small clinical trials, largely in patients with advanced blood cancers and has also shown a promising window of hope in head and neck (especially oral) cancers as well. But these treatments have nevertheless captured the attention of the people because of the remarkable responses they have produced in some patients for whom all other treatments had stopped working. The current concept of immunotherapy involves the cancer vaccines making use of CAR T-cells which are the most powerful antigen presenting cells for the induction of antigen specific T cell response. This evidence based study therefore aims to highlight the clinical perspective of CAR T-Cell based immunotherapy in oral and other head and neck cancers. Aim: Assessment of efficacy of Car T-Cell Therapy in head and neck Malignancies. Research Question: Is Car T-Cell Therapy actually effective in treating head and neck cancers? Materials and Methods: Study sample included review of 70 research articles, based on scientific data bases from the English literature based COCHRANE collaboration having a definite RCT (Randomized Control Trial). The literature was studied, analyzed and assessed; comparison was made on their p (probability) values between various techniques in terms of their sensitivity and specificity. The articles were scrutinised based on the criterion for meta-analysis and finally 11 study articles were chosen for the study. Result and Conclusion: Due to its unique individual characteristics, it helps combat against the cancer cells at its very inception. Promises a complete and permanent cure for malignancies at the grass root level. It has a sensitivity and specificity of greater than 80-90% and enhances recovery rate from 40-50% to more than 90%. So on a bulls eye view, we can say that as the already existing techniques of oral cancer treatment are very superficial and not very significant, Car T-Cell therapy tends to bring about a paradigm shift in oral cancer treatment thereby giving a new ray of hope to cancer ailing patients. Keywords: Car T-Cell Therapy, Antigen presenting cells, Cancer, Immunotherapy Introduction Cancer, the six letter word agonizing and annihilating, obliterating and destroying innocent lives since the beginning of time is a silent killer within people. Cancer, according to the World Health Organization (WHO) is defined as A large group of diseases characterized by the growth of abnormal cells beyond their usual boundaries that can then invade adjoining parts of the body and/or spread to other organs. Various conventional treatment methods have been tried so far and have had its failures and successes like Volume 3 Issue 2 1 of 5

surgical excision, radiotherapy and immunotherapy. However, these conventional techniques have started to fade giving rise to the current concept of Immunotherapy. Immunotherapy is a type of cancer treatment that boosts the body s natural defenses to fight against cancer. The current concept of immunotherapy involves Chimeric antigen receptor or the CAR T-Cell therapy which involves alterations and modifications of T cells to fight cancer cells better. Until recently, the use of CAR T-cell therapy has been restricted to small clinical trials, largely in patients with advanced blood cancers and has also shown a promising window of hope in head and neck (especially oral) cancers as well. But these treatments have nevertheless captured the attention of the people because of the remarkable responses they have produced in some patients for whom all other treatments had stopped working. The current concept of immunotherapy involves the cancer vaccines making use of CAR T-cells which are the most powerful antigen presenting cells for the induction of antigen specific T cell response. This evidence based study therefore aims to highlight the clinical perspective of CAR T-Cell based immunotherapy in oral and other head and neck cancers. Materials and Methods Study sample included review of 70 research articles, based on scientific data bases from the English literature based COCHRANE collaboration having a definite RCT (Randomized Control Trial). The literature was studied, analyzed and assessed; comparison was made on their p (probability) values between various techniques in terms of their sensitivity and specificity. The articles were scrutinised based on the criterion for meta-analysis and finally 11 study articles were chosen for the study. So when the P values or probability values of all the 11 articles were subjected to meta-analysis, we obtained a forest plot as seen in Figure 2. To interpret this forest plot, to the extreme left of the graph are the 11 articles chosen for meta-analysis and the middle of the graph represents the odds ratio and standard deviation values along with the p values of all the 11 articles chosen. Towards the extreme right, moving onto the graph proper there is a central line called the median line or the null hypothesis line. The median line has boxes on the right and left of the line signifying the sample sizes of each study that was chosen. The boxes on the left of the graph proper and the ones touching the median line states that those particular studies are not significant but the ones on the right side of the median line and not touching the line are considered to be significant. Finally, the diamond shaped box on the bottom of the graph (which is a summation of all the probability values) is neither touching the median line and is on the right side of the line proving that our study is significant. This meta-analysis conducted in order to document the significance of the Car T-Cell therapy although in the early era of acceptance has been found to be highly significant and a potent future weapon of fight against oral cancers with high sensitivity and specificity. Discussion Oral cancer is one of the 10 most common cancers in the world, with a delayed clinical detection, poor prognosis, without specific biomarkers for the disease and expensive therapeutic alternatives. The genetic engineering of T cells through the introduction of a chimeric antigen receptor (CAR) allows for generation of tumortargeted T cells. Once expressed by T cells, CARs combine antigenspecificity with T cell activation in a single fusion molecule. Most CARs are comprised of an antigen-binding domain, an extracellular spacer/hinge region, a trans-membrane domain and an intracellular signaling domain resulting in T cell activation after antigen binding. Figure 1: Schematic representation of the selection criterion for the studies in this meta-analysis Result and Conclusion Tumor associated antigens can be divided into several groups including antigens that 1. Contain novel peptide sequences due to gene mutation. 2. Are expressed in a tissue/lineage specific fashion. 3. Are normally expressed during fetal development or at immunoprivileged sites. 4. Are expressed at higher than normal levels on tumor cells compared to non-malignant host cells. Mutated Antigens Ideally, the targeted antigen should contain a novel peptide sequence, limiting its expression to tumor cells. One example is a splice variant of the epidermal growth factor receptor (EGFRvIII). Other antigens that may provide some target exclusivity include those with altered post-translational modifications or those that present conformational epitopes unique to the tumor microenvironment. Figure 2: Forest plot or meta-analysis graph of our study Tissue/Lineage Antigens Tissue/Lineage restricted antigens by definition show restrictive expression patterns and may seem particularly attractive. While tissue expendability is acceptable or correctable in certain conditions like B-cell aplasia with CD19 CART cells, the function of most solid organs cannot be readily replaced. Volume 3 Issue 2 2 of 5

Developmental Antigens Antigens expressed during fetal development or at immunoprivileged sites such MAGE family members or NY-ESO-1 is actively being targeted with αβ TCRs. However most of these antigens are in cytoplasm, making them inaccessible to scfv-based CARs that recognize antigens expressed on the cell surface. Overexpressed Antigens The majority of targeted antigens are only overexpressed in comparison to normal tissues, raising concerns about on target/ off tumor side effects, which in most cases cannot be adequately assessed in murine models. Manufacturing and Preparation of Car T-Cells sustained and adequate activation. T-cell activation needs a primary specific signal via the T-cell receptor (Signal 1) and costimulatory signals such as CD28, 4-1BB, or OX40 (Signal 2). T-cell activation is also required for the transduction of the CAR cdna via retroviral vectors. T-Cell activation can be done via : Cell-Based T-Cell Activation Antigen-presenting cells, such as dendritic cells (DCs), are the endogenous activators of T-cell responses. While therapeutic applications of DCs continue to be investigated, DC potency varies from patient to patient. Such limitation hampers the usage of DCs as a reliable source for T-cell activation. Beads-Based T-Cell Activation Several biotech companies have generated off-the-shelf clinicalgrade T-cell activation reagents including the Invitrogen CTS Dynabeads CD3/28, the Miltenyi MACS GMP ExpAct Treg beads, Miltenyi MACS GMP TransAct CD3/28 beads, and the Juno Stage Expamer technology. Figure 3: Schematic representation of manufacturing of Car T-Cells Steps Include 1. Patient blood collection at the hospitals and clinics are carried out and sent in vials of preserving mediums like Leukopacks that improve the viability of the cells. 2. The CAR-T cell-manufacturing processing starts from the collection of peripheral blood mononuclear cell from the patient, commonly achieved by a leukapheresis process. Consenting physicians choose the appropriate window for collection based on treatment regimens to ensure the presence of sufficient numbers of T lymphocytes. 3. Collected apheresis products can be processed in various ways depending on the downstream procedures. Devices such as Haemonetics Cell Saver 5+, COBE2991, and Fresenius Kabi LOVO have the ability to remove gross red blood cells and platelet contaminants. 4. Terumo Elutra and Biosafe Sepax systems provide size-based cell fractionation for the depletion of monocytesand the isolation of lymphocytes. 5. Instruments such as CliniMACS Plus and Prodigy systems allow the enrichment of specific subsets of T cells, such as CD4+, CD8+, CD25+, or CD62L+ T cells using Miltenyi beads postcell washing as shown in Figure 2. 6. The processed T-cell source material can either be used directly for downstream procedure or cryopreserved for future use. There are pros and cons for either practice. Nevertheless, cryopreserving the processed T cells allows time for product release testing and more flexibility for downstream process planning. 7. T-cell activation: The ex vivo expansion of T cells requires Antibody-Coated Magnetic Beads Dynabeads CD3/28 is uniform super-paramagnetic beads covalently coupled to CD3 and CD28 antibodies. The added value of this reagent is that it enables the selection and activation of T cells in a single step when used in conjunction with the Dynal ClinExVivo MPC magnet. Antibody-Coated Nano Beads Miltenyi MACS GMP TransAct CD3/28 beads are polymeric nanomatrix conjugated to CD3 or to CD28 monoclonal antibodies. The advantage of the TransAct CD3/CD28 beads is that they are biodegradable, and therefore do not require removal prior to formulation, although upstream T-cell purification is needed prior to activation. Expamer Technology The most recent development in T-cell activation reagent is the Expamer from Juno Therapeutics. Its unique core Streptamer technology has been used to isolate viral-specific lymphocytes. It has been reported recently that as a soluble and dissociable T-cell stimulation reagent, Expamer efficiently induces T cell receptor (TCR) signaling and efficiently activates T cells to support retroviral transduction and expansion. 8. Genetic modification of T cells: Current CAR-T cell therapies largely rely on stable CAR expression upon delivery by viral and nonviral gene transfer systems. There are three major types of stable gene expression vectors used for clinical applications: σ-retroviral vectors, lentiviral vectors, and the transposon/ transposase system. Messenger RNA transfer-mediated gene expression is another method to introduce CARs into cells while avoiding long-term expression. Both retroviral and lentiviral vectors are complicated complex biological reagents that require intensive and expensive biosafety testing. A relatively new plasmid based expression system, the transposon/transposase system, has been used to introduce anti- CD19 CAR into T cells by electroporation. The advantages of this system are its simple manufacturing procedure, relatively low cost, and straightforward release testing. Integration is random, posing a potential oncogenic risk secondary to mutagenesis. Volume 3 Issue 2 3 of 5

9. Expansion of CAR-T cells: Depending on the CAR-T cell modification strategy, there are several expansion platforms readily available to generate therapeutic doses of CAR-T cells. Expansion of CAR-T cells using GE bioreactors Expansion of CAR-T cells using G-Rex bioreactors Expansion of CAR-T cells using Prodigy Expansion of CAR-T cells through recursive AAPC stimulation 10. Finally, it is formulated and cryopreserved and sent to the clinic where it is infused back into the patient intravenously. Mechanism of Action in the Human Body CASCADE OF CAR-T CELLS Figure 4: Schematic representation of how Car T-cells act on the tumour cells in the body Mechanism of Action of Car T-Cells in the Human Body First, the tumour cells are identified. Then the patient s blood is collected and as given in FIG-2 CAR T cells are produced. Then sent to the clinics where it is intravenously infused into the patient. Where these CAR T cells travel through the blood circulation and activate to attack the tumour cells thereby reducing its size. It is usually done in two to three sessions in the hospital (Figure3). Some Toxic Effects Car T-Cell Therapy Includes Cytokine Release Syndrome (CRS) This potentially serious side effect is frequently associated with CAR T-cell therapy. Cytokines (chemical messengers that help the T cells carry out their functions) are produced when the CAR T cells multiple in the body and kill the cancer cells. CRS symptoms can range from mild flulike symptoms that include nausea, fatigue, headache, chills and fever to more serious symptoms, such as a low blood pressure, tachycardia (abnormally rapid heart rate), capillary leakage (fluid and proteins leak out of tiny blood vessels and flow into surrounding tissues, resulting in dangerously low blood pressure), cardiac arrest, cardiac arrhythmias, cardiac failure. B-Cell Aplasia CAR T-cell therapy targeting antigens found on the surface of B cells not only destroys cancerous B cells but also normal B cells. Therefore, B cell aplasia (low numbers of B cells or absent B cells) is an expected result of successful CD19-specific CAR T-cell treatment and has served as a useful indicator of ongoing CAR T-cell activity. This effect results in less ability to make the antibodies that protect against infection. Intravenous or subcutaneous immunoglobulin replacement therapy may be given with the aim of preventing infection. Tumor Lysis Syndrome (TLS) Another known side effect of CAR T-cell therapy is tumor lysis syndrome (TLS), a group of metabolic complications that can occur due to the breakdown of dying cells-usually at the onset of toxic cancer treatments. However, TLS can be delayed and may occur one month or more after CAR T-cell therapy. TLS can cause organ damage and can be a life-threatening complication of any treatment that causes breakdown of cancer cells, including CAR T cells. The complication has been managed by standard supportive therapy. Conclusion The actual need for this meta-analysis was laid on the foundation of following conclusions which were drawn from the final 11 studies that were finally selected after having undergone the inclusion and exclusion criterion, via; Due to its unique individual characteristics, it helps combat against the cancer cells at its very inception. Promises a complete and permanent cure for malignancies at the grass root level. It has a sensitivity and specificity of greater than 80-90% and enhances recovery rate from 40-50% to more than 90%. Figure 5: Summarising the production and action of Car T-Cells So on a bulls eye view, we can say that as the already existing techniques of oral cancer treatment are very superficial and not very significant, Car T-Cell therapy tends to bring about a paradigm shift in oral cancer treatment thereby giving a new ray of hope to cancer ailing patients. References 1. Curran KJ, Pegram HJ, Brentjens RJ (2012) Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. J Gene Med 14: 405-415. 2. Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ (2017) Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med 9: 1183-1197. 3. Xiuyan Wang, Isabelle Rivière (2016) Clinical manufacturing Volume 3 Issue 2 4 of 5

of CAR T cells: Foundation of a promising therapy. Mol Ther Oncolytics 3: 16015. 4. Shivani Srivastava, Stanley R Ridell (2015) Trends in Immunology. 5. Kakarla S, Gottschalk S (2014) CAR T cells for solid tumors: armed and ready to go? Cancer J 20: 151-155. 6. Kochenderfer JN, Rosenberg SA (2013) Treating B-cell cancer with T cells expressing anti-cd19 chimeric antigen receptors. Nat Rev Clin Oncol 10: 267-276. 7. Almåsbak H, Aarvak T, Vemuri MC (2016) CAR T Cell Therapy: A Game Changer in Cancer Treatment. J Immunol Res 2016: 5474602. 8. Bruce L Levine, James Miskin, Keith Wonnacott, Christopher Keir (2017) Global Manufacturing of CAR T Cell Therapy 4: 92-101. 9. Maude SL, Teachey DT, Porter DL, Grupp SA (2015) CD19- targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 125: 4017-4023. 10. Maude SL, Barrett D, Teachey DT, Grupp SA (2014) Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J 20: 119-122. Copyright: 2019 Avisesh Manohar, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Volume 3 Issue 2 5 of 5