Targeting Oncogenic Drivers

Similar documents
New Developments in Cancer Treatment. Dulcinea Quintana, MD

Early Embryonic Development

Deregulation of signal transduction and cell cycle in Cancer

BIT 120. Copy of Cancer/HIV Lecture

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis.

number Done by Corrected by Doctor Maha Shomaf

oncogenes-and- tumour-suppressor-genes)

CANCER. Inherited Cancer Syndromes. Affects 25% of US population. Kills 19% of US population (2nd largest killer after heart disease)

Chapter 4 Cellular Oncogenes ~ 4.6 -

Cancer Genetics. What is Cancer? Cancer Classification. Medical Genetics. Uncontrolled growth of cells. Not all tumors are cancerous

Multistep nature of cancer development. Cancer genes

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

What causes cancer? Physical factors (radiation, ionization) Chemical factors (carcinogens) Biological factors (virus, bacteria, parasite)

Section D: The Molecular Biology of Cancer

New Oncology Drugs: Nadeem Ikhlaque, M.D Subtitle Would Go Here

Chapter 9, Part 1: Biology of Cancer and Tumor Spread

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Development of Carcinoma Pathways

Karyotype analysis reveals transloction of chromosome 22 to 9 in CML chronic myelogenous leukemia has fusion protein Bcr-Abl

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Determination Differentiation. determinated precursor specialized cell

Lecture 8 Neoplasia II. Dr. Nabila Hamdi MD, PhD

Cancer genetics

CELL BIOLOGY - CLUTCH CH CANCER.

Transformation of Normal HMECs (Human Mammary Epithelial Cells) into Metastatic Breast Cancer Cells: Introduction - The Broad Picture:

Cancer and Gene Alterations - 1

Ch. 18 Regulation of Gene Expression

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

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

Corporate Medical Policy

The Role of Targeted Therapy in Metastatic Colorectal Cancer Patient Care

1. Basic principles 2. 6 hallmark features 3. Abnormal cell proliferation: mechanisms 4. Carcinogens: examples. Major Principles:

Backgrounder. 1. What are targeted therapies? 2. How do targeted therapies work?

Section D. Genes whose Mutation can lead to Initiation

CANCER IMMUNOTHERAPY: A REVIEW

Targeted Therapy Vijay Narang

TARGETED THERAPY FOR CHILDHOOD CANCERS

Activation of cellular proto-oncogenes to oncogenes. How was active Ras identified?

- A cancer is an uncontrolled, independent proliferation of robust, healthy cells.

Name Section Problem Set 6

VIRUSES AND CANCER Michael Lea

Personalized Genetics

Immunology CANCER IMMUNOLOGY

Precision Genetic Testing in Cancer Treatment and Prognosis

CELL CYCLE MOLECULAR BASIS OF ONCOGENESIS

Cancer Cells. It would take another 20 years and a revolution in the techniques of biological research to answer these questions.

EBV infection B cells and lymphomagenesis. Sridhar Chaganti

7.012 Problem Set 6 Solutions

New Developments in Cancer Treatment. Ian Rabinowitz MD

Third line of Defense

Genetics of Cancer Lecture 32 Cancer II. Prof. Bevin Engelward, MIT Biological Engineering Department

Molecular Cell Biology. Prof. D. Karunagaran. Department of Biotechnology. Indian Institute of Technology Madras

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

Problem Set 5 KEY

Introduction to Cancer Biology

Cancer. Questions about cancer. What is cancer? What causes unregulated cell growth? What regulates cell growth? What causes DNA damage?

BY Mrs. K.SHAILAJA., M. PHARM., LECTURER DEPT OF PHARMACY PRACTICE, SRM COLLEGE OF PHARMACY

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

Introduction to Cancer Bioinformatics and cancer biology. Anthony Gitter Cancer Bioinformatics (BMI 826/CS 838) January 20, 2015

Genome of Hepatitis B Virus. VIRAL ONCOGENE Dr. Yahwardiah Siregar, PhD Dr. Sry Suryani Widjaja, Mkes Biochemistry Department

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

Lecture 1: Carcinogenesis

Cancer immunity and immunotherapy. General principles

Drug Discovery Platform: Cancer Cell Signaling. MET Inhibitor. Merestinib, LY Christensen JG, et al1; Eder JP, et al 2

MOLECULAR BASIS OF ONCOGENESIS

609G: Concepts of Cancer Genetics and Treatments (3 credits)

Dr Rodney Itaki Lecturer Anatomical Pathology Discipline. University of Papua New Guinea School of Medicine & Health Sciences Division of Pathology

Description of Procedure or Service. Policy. Benefits Application

Chapt 15: Molecular Genetics of Cell Cycle and Cancer

Computer Science, Biology, and Biomedical Informatics (CoSBBI) Outline. Molecular Biology of Cancer AND. Goals/Expectations. David Boone 7/1/2015

New Developments in the Treatment of Colorectal Cancer. Jonathan Loree, MD, MS, FRCPC Department of Medical Oncology BC Cancer Vancouver Centre

MET Inhibitor. Merestinib, LY Drug Discovery Platform: Cancer Cell Signaling. Derived from Christensen JG, et al 1 ; Eder JP, et al.

Tumor Immunology: A Primer

Personalized Medicine: Lung Biopsy and Tumor

Introduction to Genetics

Cell Cycle and Cancer

Test Bank for Robbins and Cotran Pathologic Basis of Disease 9th Edition by Kumar

Mohammed El-Khateeb. Tumor Genetics. MGL-12 May 13 th Chapter 22 slide 1 台大農藝系遺傳學

Clinical Policy: Nivolumab (Opdivo) Reference Number: CP.PHAR.121

Regarding techniques of proteomics, there is:

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

Cancer. Death Rates for the Main Causes of Death in the U.S. Leading Sites of New Cases & Deaths (2015 estimates in the U.S.)

Oncogenes and Tumor. supressors

Tumor suppressor genes D R. S H O S S E I N I - A S L

Carcinogenesis. Carcinogenesis. 1. Basic principles 2. 6 hallmark features 3. Abnormal cell proliferation: mechanisms 4. Carcinogens: examples

Biochemistry of Cancer and Tumor Markers

VIII Curso Internacional del PIRRECV. Some molecular mechanisms of cancer

Antibody-Based Immunotherapeutic Agents for Treatment of Non-Hodgkin Lymphoma

EGFR Antibody. Necitumumab, LY , IMC-11F8. Drug Discovery Platform: Cancer Cell Signaling

Osamu Tetsu, MD, PhD Associate Professor Department of Otolaryngology-Head and Neck Surgery School of Medicine, University of California, San

Chapter 18- Oncogenes, tumor suppressors & Cancer

Test Bank for Robbins and Cotran Pathologic Basis of Disease 9th Edition by Kumar

Lecture 1: Carcinogenesis

Oncogenes and tumour suppressor genes

Agro/Ansc/Bio/Gene/Hort 305 Fall, 2017 MEDICAL GENETICS AND CANCER Chpt 24, Genetics by Brooker (lecture outline) #17

Viruses Tomasz Kordula, Ph.D.

Institute of Radiation Biology. Oncogenes and tumour suppressor genes DoReMi Course 2014

Principles of Adaptive Immunity

Colon Cancer and Hereditary Cancer Syndromes

Transcription:

Targeting Oncogenic Drivers Yujie Zhao Alex A. Adjei Department of Medicine, Roswell Park Cancer Institute, Buffalo, N.Y., USA Abstract Cancer is a genetic disease caused by a series of somatic and/or germline mutations. The roles of oncogenes and tumor suppressors in molecular origin have been well established. Targeting oncogene products has become an attractive therapeutic strategy with great clinical success, whereas tumor suppressors are considered undruggable because current technology is not able to restore tumor suppressor function in metastatic disease. Although systematic approaches to discover genetic alterations have become available to individual patients, differentiating driver from passenger mutations and identifying and validating drug targets remain challenging. Protein tyrosine kinases play crucial roles in virtually all cellular processes and possess structural features that render them druggable. Monoclonal antibodies and small-molecule inhibitors represent two major classes of targeted therapeutic agents, each possessing its own strength and weakness. Although initial successes have been achieved, targeted therapy faces many challenges that need to be addressed and hurdles to overcome. 2014 S. Karger AG, Basel Cancer is the most common human genetic disease, developing after a series of gene alterations that lead to uncontrolled growth of cells. While a small number of hereditary syndromes are directly caused by germline mutations, the vast majority of human s are driven by sequentially accumulated somatic genetic and epigenetic changes. These genetic alterations ultimately lead to acquisition of the hallmarks of malignancy comprising resistance to cell death, sustained proliferative signaling, activation of invasion and metastasis mechanisms, angiogenesis induction and replicative immortality [1]. Additionally, inheritable genetic variations also contribute to development by influencing the susceptibility to malignancy transformation [2, 3].

Genetic alterations may promote development through either constitutive activation of proto-oncogenes or loss of function of tumor suppressor genes [4, 5]. Proto-oncogenes are normal cellular genes that encode proteins involved in cell proliferation and cell-death protection. When deregulation occurs, either through changes in the protein-coding segment or by alteration of their expression levels, proto-oncogenes are converted to oncogenes, which promote development. The first oncogene isolated was from a -inducing retrovirus, the Rous sarcoma virus, which transmits sarcomas to infected chicks through incorporation of the v-src oncogene to host genomes [6]. Since the discovery of v-src, numerous other proto-oncogenes have been identified. They can be converted to oncogenes through various genetic alterations such as point mutations, deletions, duplication/amplification and chromosome translocations, as well as epigenetic alterations. Following the understanding of oncogenes, a second group of genes in tumor development, the tumor suppressor genes, also entered the center stage of research in the molecular origin of. In contrast to oncogenes, tumor suppressor genes encode proteins that prevent tumor development or progression [4]. This is usually accomplished through maintaining genome integrity and constraining cell proliferation by regulating the cell cycle, promoting apoptosis and facilitating DNA repair. The first gene recognized as a tumor suppressor gene was a gene involved in cell cycle control, the retinoblastoma (RB) gene [7 9]. Loss of RB was found to be the cause of hereditary as well as sporadic retinoblastoma. Since in general one copy of a tumor suppressor gene is sufficient to control cell proliferation, both alleles of a tumor suppressor gene have to be lost or inactivated in order to promote tumor development. This may occur through alterations such as point mutations, deletions, chromosome deletions and translocations, and epigenetic silencing [10, 11]. In addition to oncogenes and tumor suppressors, genetic materials acquired from viruses, including human papilloma virus, Epstein-Barr virus, hepatitis B virus, human T lymphotropic virus 1 and human herpes virus 8, can also promote tumor development. This usually occurs through mechanisms such as inactivation of cellular tumor suppressors or alteration of the transcription of the neighboring genes after integration into the host genome, although viral genes may function as oncogenes themselves as well [12, 13]. The understanding of the molecular origin of development has led to investigations of new therapeutic agents [14]. While strategies to restore tumor suppressor gene functions have been hindered by technical hurdles of ineffective gene delivery, numerous new therapeutics targeting oncoproteins have been evaluated in clinical studies with many approved for clinical use. Identification of Cancer Therapy Targets One of the most exciting developments in biomedical research over the past decade has been the technological advances in genome sequencing. The availability of nextgeneration sequencing (NGS) has revolutionized genome sequencing. Com- 2 Zhao Adjei

pared with previous sequencing methods, NGS executes reactions in a massively parallel fashion, and thus millions of short sequence fragments are read simultaneously, and the reaction steps occur in parallel with the detection steps. This not only allows rapid sequence reading, but also provides improved coverage and increased accuracy since the same segment of a DNA sequence is read repeatedly [15]. The dramatically reduced cost and turnaround time of NGS makes sequencing of entire genomes for each individual patient technologically and financially possible [16, 17]. With the availability of this new technology on the horizon for clinical care, the pressing question now is how to identify mutations that play causal roles in. Genetic alterations described in tumors include single-base substitutions, small insertions and deletions (indels), amplifications, homozygous deletions, and translocations. These alterations can be compiled into two categories, drivers and passengers ; while the former confer growth advantage on cells carrying them and are required to sustain proliferation of cell, the latter are not. While more than one driver is likely required to sustain tumor growth, the number of driver mutations required in each tumor type has not been well established [13]. Since is genetically unstable, a cell often bears numerous genetic alterations. The ultimate assay to distinguish drivers from passengers is the transformation assay, which however is often not feasible due to the sheer volume of mutations existing in cells. Alternatively, predictions can be made based on other factors. Whether a mutation can lead to potential functional impact due to protein coding sequence alterations may be predicated using computational tools. Mutations in genes that have been previously identified as related or involved in cellular signal transduction pathways that play significant roles in malignancy transformation are more likely to be driver mutations [16, 18, 19]. Since passenger mutations are assumed to occur randomly throughout the genome, whereas driver mutations occur frequently in related genes, a gene that exhibits higher prevalence of somatic mutations than expected by chance is considered more likely to be a driver mutation, even though it has been recognized that passenger mutations may occur in clusters, and driver mutations can exist at low frequency in cells [18, 19]. Since is an evolutionary process with continuous acquisition of new genetic alterations, additional driver mutations can be acquired after development, and clones harboring these mutations will expand under positive selection due to enhanced growth potential and/or treatment resistance [20 23]. Moreover, a preexisting passenger mutation may become a driver mutation by contributing to treatment resistance and clonal expansion [24]. In addition to identifying drivers, genome profiling-based therapy selection faces other hurdles. The discordance between primary tumor and recurrent/metastatic lesions and intra-tumor heterogeneity raise the question of whether a single biopsy sample can be used to determine the genomic landscape of the entire disease [25]. Whether the driver mutations discovered are actionable, and how to design clinical studies to match patients with targeted agents are also among the challenges remaining to be addressed [26]. Targeting Oncogenic Drivers 3

Approaches to Inhibit Driver Genetic Alterations Therapeutic targeting of driver genetic alterations has become a major focus of oncology. The development of new therapies is often based on inhibition of disease-associated molecular interactions. Modern biology has facilitated the search for new small molecules that potently and selectively modulate the functions of molecular targets via this approach. Additionally, monoclonal antibodies, which exert their antitumor activity through neutralizing and/or depleting targeted molecules or target positive cells, have also become a vital drug class in targeting -related molecular pathways [27, 28]. Other types of targeted therapeutic agents such as peptide mimetics and sirnas have also emerged and are being evaluated in clinical studies [29, 30]. Therapeutic Monoclonal Antibody The modern era of monoclonal antibody therapy began after the development of hybridoma technology in 1975, which enabled the continuous supply of specific monoclonal antibodies [31]. Among all antibody isotypes, immunoglobulin G (IgG) is the most frequently used monoclonal antibody in therapy. Structurally, it comprises an antigen binding (Fab) and a constant fragment (Fc) region. While Fab binds specific antigen, Fc interacts with IgG receptors (FcγRs) and complement, mediating antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC). Fc also interacts with neonatal FcR, which is responsible for the long serum half-life of IgG. Since murine antibodies are immunogenic, lack effector functions, and have a short half-life when used in humans, chimerization and humanization are required in order to achieve adequate efficacy [32]. Additionally, fully human antibodies obtained by screening phage display libraries expressing human antibody fragments or expressed by mice genetically engineered with human immunoglobulin gene sequences have also become available for clinical use [27, 33]. The antitumor activities of monoclonal antibodies are exerted through both immune and nonimmune mechanisms, including binding and depletion of growth factors, blockade of ligand and receptor interactions and signaling, and cellular depletion through ADCC, ADCP and CDC [27]. In addition to the antitumor effect medicated by naked antibodies, antibody-drug conjugates carrying drugs, toxins or radionucleotides have also been developed and gained approval for clinical use [34]. Since the approval of the first therapeutic monoclonal antibody rituximab in November 1997, thirteen monoclonal antibodies have been introduced to clinic for treatment of solid tumors and hematological malignancies (table 1). Currently, there are about 300 monoclonal antibodies at various stages of clinical development. Among them, 10 are undergoing evaluation in phase 2/3 or phase 3 clinical studies [35]. In general, treatments with currently approved monoclonal antibodies are better toler- 4 Zhao Adjei

Table 1. Monoclonal antibodies approved for clinical use Molecular target(s) Mechanism of action Treatment indications Cetuximab EGFR/ErbB1 mouse/human chimeric IgG1 squamous cell carcinoma of the head and neck, KRAS wild-type colorectal Panitumumab EGFR/ErbB1 human IgG2 KRAS wild-type colorectal Trastuzumab ErbB2/HER2/Neu humanized IgG1 HER2-overexpressing breast, gastric or gastroesophageal junction adenocarcinoma Pertuzumab ErbB2/HER2/Neu humanized IgG1 HER2-overexpressing breast Bevacizumab Vascular endothelial growth factor A humanized IgG1 Rituximab CD20 mouse/human chimeric IgG1 glioblastoma, colorectal, non-small cell lung, renal cell follicular, CD-20-positive, B-cell non-hodgkin lymphoma Ofatumumab CD20 human IgG1 chronic lymphocytic leukemia Alemtuzumab CD52 humanized IgG4 chronic lymphocytic leukemia Ipilimumab cytotoxic T lymphocyteassociated antigen 4 (CTLA-4) human IgG1 melanoma Tremelimumab anti-ctla-4 human IgG2 melanoma Tositumomab and iodine I 131 tositumomab CD-20 murine IgG2 iodine-131 conjugate non-hodgkin s lymphoma 90 Yttrium-ibritumomab tiuxetan CD20 murine IgG1 chelator tiuxetan conjugate Brentuximab vedotin CD30 chimeric IgG1-monomethyl auristatin E conjugate non-hodgkin s lymphoma Hodgkin lymphoma, systemic anaplastic large-cell lymphoma ated compared with conventional chemotherapeutic agents, although toxicity profiles may vary depending on the therapeutic targets, with the exception of infusion reactions, which have been reported across all agents. Skin rash and cardiac dysfunction were observed with epidermal growth factor receptor (EGFR)- and HER2-targeted agents, respectively; hypertension, thrombosis, hemorrhage and delayed wound healing were observed with vascular endothelial growth factor-targeted agents; infection and cytopenia are more commonly associated with monoclonal antibodies targeting hematological malignancies, while immune-related adverse events have been reported with immune-modulating agents. Most of the monoclonal antibodies approved for Targeting Oncogenic Drivers 5