Harnessing Complexity: Companion Diagnostics in Oncology

Similar documents
Point of View Harnessing Complexity: Companion Diagnostics in Oncology

Transform genomic data into real-life results

UNDERSTANDING SQUAMOUS CELL LUNG CANCER

Molecular Testing Updates. Karen Rasmussen, PhD, FACMG Clinical Molecular Genetics Spectrum Medical Group, Pathology Division Portland, Maine

Patient Leader Education Summit. Precision Medicine: Today and Tomorrow March 31, 2017

7/6/2015. Cancer Related Deaths: United States. Management of NSCLC TODAY. Emerging mutations as predictive biomarkers in lung cancer: Overview

GENETIC TESTING FOR TARGETED THERAPY FOR NON-SMALL CELL LUNG CANCER (NSCLC)

Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment with Targeted Tyrosine Kinase Inhibitors

1. Q: What has changed from the draft recommendations posted for public comment in November/December 2011?

Role of the pathologist in the diagnosis and mutational analysis of lung cancer Professor J R Gosney

TARGETED THERAPY FOR LUNG CANCER. Patient and Caregiver Guide

Consensus statement between CM-Path, CRUK and the PHG Foundation following on from the Liquid Biopsy workshop on the 8th March 2018

Regulatory Landscape for Precision Medicine

Molecular Testing in Lung Cancer

Corporate Medical Policy

Product Introduction

Delivering on the Promise of Personalised Healthcare

Disruptive innovation in molecular diagnostics. Hilde Windels CEO Biocartis 25 March 2017

MICROSCOPY PREDICTIVE PROFILING

Tissue or Liquid Biopsy? ~For Diagnosis, Monitoring and Early detection of Resistance~

Personalised Healthcare (PHC) with Foundation Medicine (FMI) Fatma Elçin KINIKLI, FMI Turkey, Science Leader

QIAGEN Complete Solutions for Liquid Biopsy Molecular Testing

Learning from the Impact of the Drug-Diagnostics Strategy in Oncology

Advances in Pathology and molecular biology of lung cancer. Lukas Bubendorf Pathologie

The Expanding Value of Biomarkers in NSCLC Treatment

Personalized Genetics

IRESSA (Gefitinib) The Journey. Anne De Bock Portfolio Leader, Oncology/Infection European Regulatory Affairs AstraZeneca

Personalised Healthcare. will it deliver? Bob Holland Head of Personalised Healthcare & Biomarkers Innovative Medicines AstraZeneca R&D

Delivering Value Through Personalized Medicine: An Industry Perspective

Accelerate Your Research with Conversant Bio

Review of NEO Testing Platforms. Lawrence M. Weiss, MD Medical Director, Aliso Viejo

Companion & Complementary Diagnostics: Clinical and Regulatory Perspectives

NCCN Non-Small Cell Lung Cancer V Meeting June 15, 2018

PD-L1 Analyte Control DR

This online (electronic) survey contains twenty four simple questions. Those marked with an asterisk (*) must be answered.

Biomarcatori per la immunoterapia: cosa e come cercare Paolo Graziano

MP Circulating Tumor DNA Management of Non-Small-Cell Lung Cancer (Liquid Biopsy)

Agilent companion diagnostics for cancer immunotherapy

Personalized Medicine: Lung Biopsy and Tumor

Performance Improvement Strategies in Non-Small Cell Lung Cancer Audioconference - April 22, 2010

Jules Bordet Institute, Brussels, Belgium Université Libre de Bruxelles Breast International Group (BIG aisbl), Chair ESMO President

HOW TO MAXIMIZE PATIENT RECRUITMENT IN ONCOLOGY TRIALS A BIOPHARMA DIVE PLAYBOOK

HER2 status assessment in breast cancer. Marc van de Vijver Academic Medical Centre (AMC), Amsterdam

Implementation of nation-wide molecular testing in oncology in the French Health care system : quality assurance issues & challenges

Evolution of Pathology

Personalized Healthcare Update

Introduction to liquid biopsies. Rachel Butler All Wales Genetics Laboratory

Circulating Tumor DNA for Management of Non-Small-Cell Lung Cancer (Liquid Biopsy)

Circulating Tumor DNA in GIST and its Implications on Treatment

Non-Profit Startup Paradigm Launches Cancer Panel Based on DNA, RNA Sequencing

Integration of Genomics Into Clinical Pathways. Precision Medicine and Decision Support

Precision Genetic Testing in Cancer Treatment and Prognosis

CDx in oncology Prof. Christophe Le Tourneau, MD, PhD FEAM Geneva September 27, 2018

Overview of Biomarker Development for Immune PD-1/L1 Checkpoint Blockade

Predictive markers for treatment with Immune checkpoint inhibitors - PD-L1 et al -

Joachim Aerts Erasmus MC Rotterdam, Netherlands. Drawing the map: molecular characterization of NSCLC

Erlotinib for the first-line treatment of EGFR-TK mutation positive non-small cell lung cancer

Test Category: Prognostic and Predictive. Clinical Scenario

Pathology Report Patient Companion Guide

How could molecular profiling impact histology independent labels in the future?

Educator Navigation Guide

Medical Policy An independent licensee of the Blue Cross Blue Shield Association

Diagnostics guidance Published: 14 August 2013 nice.org.uk/guidance/dg9

Comprehensive Genomic Profiling, in record time. Accurate. Clinically Proven. Fast.

MATCHMAKING IN ONCOLOGY CHALLENGES AND COMBINATION STRATEGY FOR NOVEL TARGETED AGENTS

UCL-Advanced Diagnostics. 2015/16 Service Update

Personalized Therapies for Lung Cancer. Questions & Answers

Technology appraisal guidance Published: 16 May 2018 nice.org.uk/guidance/ta520

Guideline. Associated Documents ASCO CAP 2018 GUIDELINES and SUPPLEMENTS -

GPS Cancer. The Era of Complete Genomics and Proteomics is Here. Advanced molecular profiling to inform personalized treatment strategies

REFERENCE CODE GDHC196DFR PUBLICAT ION DATE JUNE 2013 ABRAXANE (NON-SMALL CELL LUNG CANCER) FORECAST AND MARKET ANALYSIS TO 2022

GLOBAL REGISTRATION STRATEGIES:

Innovation, Uncertainty and Reimbursement Processes in Precision Medicine: The Case of PD-L1

The Role of Immuno-Oncology Biomarkers in Lung Cancer

REFERENCE CODE GDHC207DFR PUBLICAT ION DATE JUNE 2013 GILOTRIF (NON-SMALL CELL LUNG CANCER) FORECAST AND MARKET ANALYSIS TO 2022

REFERENCE CODE GDHC198DFR PUBLICAT ION DATE JUNE 2013 TARCEVA (NON-SMALL CELL LUNG CANCER) FORECAST AND MARKET ANALYSIS TO 2022

Diagnostic test Suggested website label Description Hospitals available

ECMC cfdna consensus meeting

BREAST CANCER PATHOLOGY

StrandAdvantage Tissue-Specific Cancer Genomic Tests. Empowering Crucial First-Line Therapy Decisions for Your Patient

Data Sharing Consortiums and Large Datasets to Inform Cancer Diagnosis

CANCER. Clinical Validation of Breast Cancer Predictive Markers

FDA APPROVES TARCEVA (ERLOTINIB) TABLETS AND COBAS EGFR MUTATION TEST FOR SPECIFIC TYPE OF LUNG CANCER

Reflex Testing Guidelines for Immunotherapy in Non-Small Cell Lung Cancer

Role of the Pathologist in Guiding Immuno-oncological Therapies. Scott Rodig MD, PhD

QIAGEN's Growing Immuno-Oncology Testing Portfolio

Opportunities and Challenges in the Development of Companion Diagnostics

MEDICAL POLICY. Proprietary Information of YourCare Health Plan

Media Release. Basel, 26 March 2018

Quality in Control. ROS1 Analyte Control. Product Codes: HCL022, HCL023 and HCL024

Your Guide to the Breast Cancer Pathology. Report. Key Questions. Here are important questions to be sure you understand, with your doctor s help:

IMMUNOTHERAPY FOR LUNG CANCER

SUBJECT: GENOTYPING - EPIDERMAL GROWTH

Changing demographics of smoking and its effects during therapy

MET skipping mutation, EGFR

Read, Interpret, and Communicate Test Results

IMMUNOTHERAPY FOR THE TREATMENT OF LUNG CANCER

Making a plan can help you be confident about your journey with EGFR+ lung cancer.

immunotherapy a guide for the patient

Robert Beer

Transcription:

Ipsos Healthcare Harnessing Complexity: Companion Diagnostics in Oncology Pieter De Richter, Head of Global Molecular Diagnostics Portfolio, Ipsos Healthcare First Published: October 2015 Updated: February 2017

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY Introduction: beyond the microscope Companion diagnostics have had a profound impact on the way in which cancer is treated. As an increasing number of drugs gain approval for patients whose cancers have certain molecular characteristics, the promise of truly personalised medicine is edging closer towards reality. The realisation that every patient s cancer is different and that these differences can be detected with in vitro diagnostic tests and exploited with tailored therapies has led to a rapid fragmentation of the anticancer drug landscape. This, in turn, has led to a significantly more complicated decision-making process for oncologists in many cases. No sub-classification; all patients treated the same Classification by stage; two distinct groups of patients with different treatment approach Classification by stage and histology; four different groups of patients Classification by stage, histology and one molecular marker; 7 different groups of patients Classification by stage, histology and two molecular markers; many different groups of patients Fig 1: Symbolic representation of the increasing complexity resulting from increased segmentation of patient groups, based on non-molecular and molecular markers. Treatment decisions are now often based on the combination of those criteria, and the increasing number of biomarkers is segmenting the patient pool into smaller and smaller sub-groups Molecular diagnostics testing is at the far end of a long and evolving spectrum of narrowing down the nature of a disease. This spectrum starts with the clinical observation of (often visual) symptoms, moves on to more advanced visualization techniques involving x-rays and other imagining machinery, pulls in pathologists and their microscopes to examine the overall cell structure, involves dyes and stains to quantify the expression of certain markers on cell membranes, and finally moves deep within the cell nucleus to look for specific changes in the cancer s DNA. 3

IPSOS HEALTHCARE It is no longer sufficient to classify a patient s cancer as being an adenocarcinoma of the lung; rather, it is now crucial to know if that particular patient s adenocarcinoma of the lung has certain mutations in the gene that encodes the EGF receptor and, if so, whether those mutations are sensitising mutations as opposed to resistance mutations. This is not something that an oncologist can assess by going through the patient s symptoms in detail. Neither is it something that a radiologist can see on an X-ray, nor can it be seen by the surgeon during a biopsy, nor even the pathologist upon examination of the biopsy material under a microscope. DNA changes occur at the molecular level, and can only be detected with sophisticated machinery that rely on such advanced techniques as real time polymerase chain reactions (PCR) and, increasingly, full gene sequencing. Diagnostic imaging Whole organ Biopsy Tissue sample Histological classification Cellular structure / staining Mutational analysis DNA Fig 2: Macroscopic to sub-microscopic diagnosis and classification of lung cancer (simplified) Despite the obvious advantage of identifying patients for whom specific targeted therapies are most likely to lead to a good response, the growing number of companion diagnostics is causing ever-increasing levels of complexity. This has potentially profound effects, from hampering the uptake of certain drugs to patients not receiving therapies that could be of benefit to them. It is this conundrum, the delicate balance between companion diagnostics clinical utility and their potential confounding impact on patient treatment, that this article has as its central theme.

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY Predicting response: perfection or educated guesswork? The first question concerning companion diagnostics is perhaps the biggest one of all: do they increase a drug s usage due to its demonstrably superior effects in a specific patient population, or do they limit a drug s uptake due to the exclusion of (often large) groups of patients who lack the required biomarker status? In order to answer this complex question, we must first embark upon a scientific journey to better understand what exactly it is a companion diagnostics test is measuring, starting with immunohistochemistry testing for HER2. Breast cancer cells, like most high-incidence tumours, are a heterogeneous lot. Apart from the histology, which distinguishes between ductal, lobular and other cell lines, we ve known for some years now that there are a number of surface proteins that are expressed to varying degrees on the cell membrane of these cancer cells. Three cell surface receptors in particular stand out: the oestrogen receptor (ER), progesterone receptor (PgR) and human epidermal growth factor receptor 2 (HER2). Whereas the hormone receptors are overexpressed by the majority of breast cancer cells, the HER2 receptor is said to vbe overexpressed by around 25% of breast cancers. So what exactly does this mean? The key point to realise is that HER2 expression (and protein expression in general) is not a binary, black-and-white parameter. The majority of cells not just breast cancer cells express HER2 to some extent; it is the exact levels of expression that matter. Normal breast tissue, and breast cancers that do not overexpress HER2, typically have 20,000 or fewer HER2 receptors per cell 1. Some breast cancers, which are said to be strongly HER2-positive, may express more than 2,000,000 receptors per cell a hundred-fold increase. Importantly, however, many breast cancer cells fall somewhere in between these two extremes. Immunohistochemistry (IHC) tests for HER2 help pathologists to assess the levels of HER2 expression (the stain in the test kits will show up more strongly the higher the concentration of receptors) and subdivide them into 4 categories but even within those categories, there is significant variation. 5

IPSOS HEALTHCARE So what does this mean from a HER2-targeted drug point of view? The answer, again, is complex. Several of the key early trastuzumab trials clearly showed a correlation between HER2 overexpression levels as measured by IHC, and the response rates to this HER2-targeted therapy. Importantly, the response rates were significantly higher in the IHC 3+ group, demonstrating that the cells that express the largest number of HER2 receptors are most likely to respond to HER2- targeted drugs 2. However, some IHC 2+ tumours were also shown to respond to trastuzumab 3 ; protein overexpression is certainly not an absolute marker for predicting response in this case. Researchers also investigated the impact of gene amplification, which is a measure of how many copies of the gene are present in the cell s DNA. A technique called fluoresence in-situ hybridisation, or FISH, is used to determine this: cancers are either said to be FISH+ (more than the usual number of copies present) or FISH- (usual number of copies present) for a specific gene, though in reality this is also a sliding scale. Often, as in the case of HER2, protein overexpression and gene amplication are closely correlated; on a cellular level, gene amplification (too many copies of a particular gene) usually leads to protein overexpression, as all those extra copies make extra proteins. Hence, nearly all IHC 3+ cancers are FISH+ for HER2, and nearly all IHC 0 or IHC 1+ cells are FISH-. The interesting group of cancers are the IHC 2+ ones, which can turn out to be either FISH- (showing a low response to HER2-targeted therapies) or FISH+ (showing a much better response to HER2-targeted therapies) 3. Therefore, FISH testing is recommended for all HER2 IHC 2+ cancers before a treatment decision is made. Interestingly, however, more recent analyses have shown that even some FISH- patients may be able to benefit from trastuzumab therapy; this means that even when using both IHC and FISH testing to guide treatment decisions, some women may miss out on treatment that could potentially benefit them1. In summary, while HER2 testing via different techniques is a powerful tool to predict treatment response to targeted therapies, linking HER2 protein expression and HER2 gene amplification to predicted outcome is by no means an exact science. What about genetic mutations? Mutations are essentially binary events: either the mutation is present or it is not. So are mutations absolute predictors of treatment response? It turns out that this is far from the case. Let s take EGFR mutations in NSCLC as an example. It has been known for some time now that the presence of certain somatic mutations in NSCLC tissue,

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY Healthy breast cell or breast cancer cell, IHC 0 Breast cancer cell, HER2 IHC 1+ Breast cancer cell, HER2 IHC 2+ Breast cancer cell, HER2 IHC 3+ Protein Expression HER2 receptors <20,000 / cell HER2 receptors ~20,000 100,000 / cell HER2 receptors ~100,000-500,000 / cell HER2 receptors 500,000+ / cell IHC examination No membrane staining or faint staining in <10% of cells Faint membrane staining in >10% of cells Weak/moderate staining in >10% of cells or complete staining in <10% Complete / intense membrane staining Predicted treatment response Low response rate to HER2-inhibitors Med response rate to HER2-inhibitors High response rate to HER2-inhibitors Correlation with gene amplification Likely to be FISH- (2 copies of HER2 gene/cell) Some are FISH-, some are FISH+ Likely to be FISH+ (>2 copies of HER2 gene/cell) Fig 3. Relationship between HER2 protein expression, IHC test results, predicted treatment response and HER2 gene amplification. such as exon 19 deletions, are associated with a better response to EGFR tyrosine kinase-inhibitors (TKIs) 5, such as erlotinib and gefitinib. These mutations are oncogenic: they do not (generally) appear in healthy tissue, which is said to be wild type for the EGFR gene. Just like healthy cells, many NSCL cancers are also wild type EGFR, while a significant subset (15-35%, depending on cell type, ethnicity, gender, smoking history, etc.) do display one or more of the aforementioned EGFR mutations. As studies have shown, EGFR TKIs are more likely to lead to a response in patients whose tumours exhibit one or more of these so-called sensitising mutations. Some of these tumours will go on to develop resistance mutations, such as T790M, which helps the cancer cells overcome the effect of the EGFR inhibitors. This is one of the reasons why not all EGFR-mutated NSCLC patients respond to EGFR TKIs. However, the reverse is also true: a significant proportion of EGFR wild-type tumours nevertheless respond to EGFR TKI treatment 6. 7

IPSOS HEALTHCARE Clinical utility of testing: a delicate balance As both the trastuzumab and gefitinib example have illustrated, using companion diagnostics to determine treatment choice may sometimes lead to a certain subset of patients not receiving potentially beneficial drugs. So why are those tests being used in the first place? While there is no universal consensus, a large proportion of oncologists and pathologists agree that these tests have real clinical utility (are accurate predictors of treatment response) and there exists a large amount of clinical Fig 4. Attribute associations with different biomarker tests amongst oncologists and pathologists in the US. The percentages shown denote the proportion of respondents associating each test with that specific attribute. The closer the attribute is to the outer rim of the chart, the more important the attribute is in making a decision whether to use a test or not. Source: Ipsos Healthcare MDx Monitor, US, 2015 Q1/Q2 (research conducted online March May) HER2 testing by IHC 56% 58% 62% 65% 69% Oncologists (N = 72) Turn-around time Cost / ease of reimbursement Ease of interpretation Large amount of supporting clinical data Accurate predictor of treatment response EGFR mutation testing 25% 44% 65% 64% 65% Turn-around time Cost / ease of reimbursement Ease of interpretation Large amount of supporting clinical data Accurate predictor of treatment response 60% 60% 71%73% 73% Pathologists (N = 55) 36% 44% 47% 62% 71% Turn-around time Cost / ease of reimbursement Ease of interpretation Large amount of supporting clinical data Accurate predictor of treatment response Turn-around time Cost / ease of reimbursement Ease of interpretation Large amount of supporting clinical data Accurate predictor of treatment response data supporting this clinical utility. This is in spite of the factors discussed earlier; in other words, a marker does not have to be perfect for it to be perceived as useful for making treatment decisions. In many doctors minds, the fact that some patients may miss out on a drug that could potentially work for them is more than compensated for by the fact that the patients they are prescribing the drug to are more likely to benefit as a result of their biomarker status. Targeted therapies carry a significant

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY financial cost, and sometimes come with potential side-effects. Having a tool (biomarker) to help make a benefit vs. cost assessment is generally seen as a positive attribute. It should be pointed out that, ultimately, this type of assessment is carried out at a much higher level by authorities such as the FDA when it comes to making a decision on drug approval. Based on all the available clinical trial data to date including response rates in specific sub-types the approval bodies must decide whether it is better for the drug to be approved for all patients (and hence risk additional expenses/toxicities from these drugs in patient groups that have low probability of gaining a benefit) or only in patients with certain mutations/overexpression profiles (and hence risk excluding a potentially large percentage of the population for whom a small, but non-zero, likelihood of benefit exists). In all this, it should be noted that the relative importance of each of these factors in this intricate balancing act can be very different depending on whose point of view you consider: the patient, the physician, the drug manufacturer, the payer or the government. In the example shown in figure 5, there are a number of factors that have a clear impact on which way the balance of an allpatients approval vs. a biomarker-specific approval will shift: 1. The proportion of patients who are biomarker high vs. biomarker moderate/low (as represented by the orange vs. yellow/grey patients 2. The proportion of patients with biomarker low status who benefit from the drug despite being biomarker low (as represented by the two grey patients who benefit vs. the six who don t) 3. The proportion of patients with biomarker moderate status who do not benefit from the drug despite being biomarker moderate (as represented by the three yellow patients who don t benefit vs. the four who do) This is, however, a very simplified view. In reality, a number of additional factors also play an often highly significant role: 1. The cost of the targeted therapy, i.e. how much money is wasted on patients who do not benefit from the drug if it receives a blanket approval? 2. The cost of testing, i.e. how much money is spent on finding out biomarker status if the drug receives a restricted approval? Note that this is not just about financial cost of the test itself, but also laboratory resources, time spent waiting for the test results, potential extra biopsies required, etc. 9

IPSOS HEALTHCARE 3. The likelihood of toxicity, i.e. how likely are patients who do not benefit from the drug to develop unnecessary toxicities? 4. The duration of response / efficacy of the drug, i.e. how much incremental benefit does the drug provide in patients who do benefit, and does this differ by biomarker status? In other words, it s not just about response rates, but also PFS / OS improvements Biomarker absent Biomarker moderate Biomarker high Low likelihood of response Moderate likelihood of response High likelihood of response Scenario 1: Drug approved for all patients No benefit from drug; financial and potential toxicity burden that could have been prevented with biomarker approval Benefit from drug; would not have received drug if biomarker approved Scenario 2: Drug approved for biomarkerhigh patients only Would not have benefited from drug; financial and potential toxicity burden avoided through use of biomarker Missing out on benefit from drug as not approved for this population No benefit from drug; financial and potential toxicity burden that could have been prevented with biomarker approval depending on cut-off used Would not have benefited from drug; financial and potential toxicity burden avoided through use of biomarker Benefit from drug; may not have received drug if biomarker approved, depending on cut-off used Missing out on benefit from drug as not approved for this population No benefit from drug; financial and potential toxicity burden that would not have been prevented even if biomarker approved No benefit from drug; financial and potential toxicity burden not avoided despite approval of biomarker Benefit from drug; would have also received drug if biomarker approved Benefit from drug; may also have received drug if approved for all patients Fig 5. Simplified representation of assessing the relative pros and cons of a drug being approved for all patients vs for biomarker-high patients only.

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY This overarching question whether a drug should be approved for patients who exhibit a specific biomarker status only, even if it has been shown to (sometimes) work for those who don t has perhaps never seemed more important than right now, with the advent of immuno-oncology agents. The PD-1 and PD-L1-inhibitors, in particular, have shown great promise for NSCLC patients whose cancers exhibit overexpression of PD-L1. However, much like in the HER2 example, PD-L1 expression is very much a sliding scale and, importantly, even some patients with low PD-L1 expression have been shown to benefit. An extra complicating factor is that there is currently no consensus on how to define PD-L1 overexpression, a factor which we will address in further depth in the next section. Different toolkits: how you measure matters We saw in the previous section that, not surprisingly, not all markers are the same. Testing for HER2 overexpression is important in breast cancer (even if, as we saw, it does not give us an ideal picture), whereas testing for the same protein in lung cancer has very limited clinical utility (at least at present). Conversely, testing for EGFR mutations in breast cancer is currently not recommended; it is not seen as an actionable mutation. In short, what doctors test for matters. The story does not end there, however. For many of these markers, there are many different ways to measure the level, presence or absence of the same marker using different approaches. Previously, we alluded to the difference between IHC and FISH for HER2 testing: the former measures the level of protein expression, the latter measures the level of gene amplification. As HER2 genes encode HER2 proteins, it is no surprise that there is a strong correlation between the two; generally speaking, the more strongly the gene is amplified, the higher the resultant protein levels will be. However, (1) this correlation is not absolute and (2) positivity by FISH has been shown to be a more direct way to predict treatment response to HER2 therapy than positivity by IHC. This is why, in those cases where a tumour has a borderline HER2 IHC score of 2+, it is the FISH result that determines the most appropriate treatment choice. Simply testing with IHC tests, which are cheaper and faster than FISH tests, is not always sufficient for making the most informed choice. 11

IPSOS HEALTHCARE On a more fundamental level, there are even different ways of testing for the presence or absence of specific mutations. The exon 19 deletions in the EGFR gene, and related mutations mentioned earlier are often tested through the use of RealTime PCR-based assays. However, sequencing techniques such as Sanger sequencing, pyrosequencing and increasingly Next Generation Sequencing (NGS) can also be used. There are also a growing number of variations on the PCR theme, including digital droplet PCR and PNA-mediated PCR clamping. Going into the exact differences between all these techniques is beyond the scope of this paper. One key difference between PCR and sequencing is that the former techniques are only able to look for a list of pre-determined mutations (e.g. T790M, L588R, etc.), whereas the latter can (in theory) sequence the whole gene, flagging any and all mutations (including rare ones, and those of uncertain significance). Next Generation Sequencing panels often include a wide range of different genes, so doctors can potentially get a report of a list of different mutations across a series of different genes, as opposed to a simple yes/no status for a limited number of specific mutations in one specific gene. Due to speed and efficiency advantages, as well as a continued reduction in cost, the use of NGS is on the increase; it is, however, still significantly lower than the use of more traditional, PCR-based assays. US MDx Monitor, EGFR mutation testing, 2015 Q1/Q2 (n=95 EGFRm-tested tissue samples) 7% 7% 14% 14% 6% 6% Next Generation Sequencing Pyrosequencing RealTime PCR 73% 73% Other (Sanger, HRM, Fragment,..) Fig 6: EGFR mutation testing methodology shares Source: Ipsos Healthcare Oncology MDx Monitor, US, 2015 Q1/Q2 (research conducted online March May 2015)

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY For any given methodology, there is still a choice of different tests for the same marker. Going back to our earlier example of EGFR mutation testing, there are several different PCR-based tests available. These include: Roche cobas EGFR Mutation Test, Qiagen therascreen EGFR RGQ PCR Kit, several other branded kits, as well as a limitless number of laboratorydeveloped/home-brew kits. The Roche cobas assay detects 41 specific mutations 7, the Qiagen therascreen assay detects 29 mutations 8 ; the former has been approved by the FDA as a companion diagnostic for erlotinib 9, whereas the latter was approved to select patients for treatment with gefinitib and gilotrif 10. This differential approval, where similar tests are approved alongside specific and different drugs, is an important point which we will come back to later. It is also important to note that doctors can, and in many cases will, make their treatment decisions based on non-fda-approved, lab-developed tests, such as the chart below illustrates: US MDx Monitor, EGFR mutation testing, 2015 Q1/Q2 (n=68 EGFRm-tested tissue samples) 16% 16% 38% 38% Home-brew / Labdeveloped test Roche cobas 22% Qiagen therascreen Other brands 24% 24% Fig 7: EGFR mutation testing brand shares Source: Ipsos Healthcare Oncology MDx Monitor, US, 2015 Q1/Q2 (research conducted online March May 2015) The availability of different test kits allows for greater flexibility and means laboratories are less limited by the exact type or brand of hardware (e.g. PCR machines) that they have at their disposal. However, the lack of standardisation can sometimes be a potential concern. For the example above, several studies have shown a very close (though not 100%) concordance between test results from the Roche cobas and the Qiagen therascreen assay, as well as Sanger sequencing. Apart from the 13

IPSOS HEALTHCARE fact that some mutations are not covered by both tests, they will almost always return the same test results when they are used to test the same sample, i.e. they are functionally interchangeable. This is not always the case, however, and this is becoming a particularly important issue for the expanding suite of PD-L1 expression tests. The following are the major commercially available test kits that have been FDA-approved for measuring PD-L1 expression: Dako PD-L1 IHC 28-8 pharmdx: Approved as a complementary diagnostic in previously treated metastatic NSCLC, with the level of PD-L1 expression to be used as an additional tool for physicians to help determine which patients will most benefit from treatment with Opdivo (nivolumab) 11 Also approved as a complementary diagnostic for Opdivo in melanoma 12 Dako PD-L1 IHC 22C3 pharmdx: Approved as a companion diagnostic in NSCLC for first line metastatic patients, with 50% tumour cells showing staining defined as PD-L1 positive for the purposes of determining eligibility for treatment with Keytruda (pembrolizumab) 13 Also approved as a companion diagnostic in NSCLC for previously treated metastatic patients, with 1% tumour cells showing staining defined as PD-L1 positive for the purposes of determining eligibility for treatment with Keytruda Ventana PD-L1 (SP142) Assay: Approved as a complementary diagnostic in metastatic urothelial cancer, to help physicians determine which patients will most benefit from treatment with Tecentriq (atezolizumab). Unlike the Dako assays, the Ventana kit uses immune cell staining, rather than tumour cell staining only, to determine PD-L1 expression levels. As is clear from the above, the various test kits while measuring the same protein biomarker differ from each other in several fundamental ways and are therefore not necessarily interchangeable. Furthermore, sensitivities/specificities may differ across kits and antibodies too, potentially resulting in weaker or stronger actual staining.

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY At the time the first version of this article was prepared for publication (October 2015), the FDA had just approved Keytruda to be used for second-plus line advanced NSCLC patients whose tumours overexpress PD-L1, as determined by the PD-L1 Dako IHC 22C3 pharmdx companion diagnostic, with a cut-off of 50%. Soon thereafter, Opdivo was approved for this patient population, regardless of PD-L1 expression levels; however, the FDA noted that the level of PD-L1 expression in NSCLC tumours may help identify patients who are more likely to benefit, and approved the PD-L1 IHC 28-8 pharmdx test alongside the drug (hence known as a complementary, rather than a companion, diagnostic). Since then, a number of interesting developments have taken place in this space, including the expanded label for Keytruda in the NSCLC setting in October 2016, which meant that (1) the existing cut-off at second line was revised to 1% and (2) the drug label now included a different cut-off at 1st vs 2nd line (50% and 1%, respectively). In the meantime, the approval of Tecentriq and its immune cell-based assay further complicated the PD-L1 expression testing story. Back in late 2015, we note that it would be interesting to see how future testing guidelines would evolve, and whether they would provide some degree of standardisation against the very complex backdrop. If anything, PD-L1 expression testing has become more complex and less standardised since then, with an array of companion and complementary diagnostics, different cut-off points, different cell types measured, and different testing requirements depending on the line of therapy. Given the number of new approvals that do not require PD-L1 expression status to be known, it could be argued that this particular biomarker has decreased in importance since its first approval as a companion diagnostic. However, the upward trend in PD-L1 expression testing rates in both NSCLC and melanoma since 2015 contradicts this; even though testing is technically not required, we do see a significant proportion of NSCLC patients being tested prior to Opdivo prescription, as well as melanoma patients prior to Keytruda prescription. 14 As could perhaps be expected based on the speed of change in this market and the rapid growth in the number of tests, drugs and indication, oncologists and pathologists do not always strictly follow the labels when it comes to matching assays to drugs. As the data below illustrates (Table 1.), a significant percentage of patients on Opdivo had their cancers tested for PD-L1 expression with the 22C3 assay (Keytruda s companion diagnostic), and some Keytruda patients were tested with home-brew test kits. 15

IPSOS HEALTHCARE Current PD-1-inhibitor Opdivo (n=32) Keytruda (n=63) Home-brew/lab-developed test 10% 24% Dako IHC 22C3 pharmdx 75% 67% Dako IHC 28-8 pharmdx 9% 9% Ventana PD-L1 SP142 assay 6% -% Table 1. PD-L1 test kit use by current PD-1 inhibitor Base: All PD-L1 tested NSCLC patients (reported by oncologists) with known test kit Source: Ipsos Healthcare PD-L1 Testing Monitor, US, 2016 Q4 (research conducted online Oct Dec 2016) The future: accelerating change and multiple paradigm shifts Despite the increasing fragmentation and growing number of clinically meaningful markers, companion diagnostics testing in oncology (and indeed, in medicine in general) is still in its infancy. HER2 testing has been around for nearly 20 years, and hormone receptor testing started many years earlier. Since then, the fundamental principles haven t changed much: tissues are extracted from solid tumours, sent to a lab, analysed with single assays or at the most, small panels and results are fed back to physicians, who then make a decision based on a small number of actionable proteins or mutations. Even in adenocarcinoma lung cancers, the number of different molecular tumour subtypes remained relatively small prior to the approval of the PD-1 inhibitors: EGFR wild type, ALK wild type: treat with chemo or chemo + Avastin EGFR mutated: treat with an EGFR-TKI EGFR wild type, ALK mutated: treat with an ALK-inhibitor

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY Since the advent of PD-L1 expression testing, and with the approval of Xalkori (crizotinib) for ROS1-mutated tumours, the number of patient types has certainly grown; even so, the number of different iterations of the various biomarkers is still very limited compared to the true complexity of the genetic landscape involved in oncogenesis The state of molecular diagnostics in oncology cannot yet be considered as true personalised medicine. However, this is all about to change. This transition period of single-marker testing is a necessary, but likely brief, stepping stone towards mass profiling/sequencing of large numbers of genes, and making treatment decisions based on patients detailed genetic profiles. Next Generation Sequencing, alluded to in the earlier sections, has been gradually increasing in use over the past few years, and is getting close to being widely adopted. Also known as massively parallel sequencing or highthroughput sequencing, it allows for many different genes often in the hundreds to be sequenced in one run, making it much more efficient than PCR-based techniques or Sanger sequencing. There have been two major barriers to widespread adoption of NGS in the past: (1) its cost and (2) its clinical utility. The first barrier is rapidly disappearing, as the cost of NGS is decreasing significantly each month. The second barrier is harder to address, and comes down to the fact that many of the genes profiled through NGS panels have not yet been extensively studied in clinical trials, leaving doctors unsure as to what to do with all that data (i.e. if there is a mutation in gene X but this mutation has not been shown to correlate with a better or worse response to any therapies, what is the clinical utility of this information?). There are other revolutions taking place in the Oncology CDx testing market. For example, the increase in so-called liquid biopsies, which utilise blood or urine samples for testing biomarkers such as EGFR mutations, is a significant new development. Tissue biopsies are invasive, time-consuming and sometimes risky, and hence patients do not usually get re-tested to see if their biomarker status has changed; being able to do so through a simple blood draw or urine collection may significantly increase re-testing rates. On the one hand, this has the potential to make things a lot easier for both doctor and patient; on the other hand, the increase of re-testing, the tracking of biomarker status as a dynamic rather than a static factor, and the choice between solid and liquid samples add further layers of complexity to this ever-changing field. 17

IPSOS HEALTHCARE Conclusion: from complexity to opportunity Cancer is, ultimately, a genetic disease: it is always caused by genetic changes. We cannot hope to conquer cancer unless we fully understand the intricate relationship between the myriad genes and proteins that control the cell cycle and which, through random mutations, can and do lead to malignancies. As we uncover more and more of these genes and proteins, we are starting to realise that they hold the key to successful treatment: no two cancers are the same, and their subtle differences can only be identified by probing deep into their sub-microscopic worlds. The tools that enable us to do so are constantly evolving, allowing us to map those molecular differences more efficiently, in much larger numbers, less invasively, and more accurately. It is easy to become overwhelmed by this flood of new data, new techniques, new methodologies, new targets, and even entirely new paradigms. This is a challenge not just faced by oncologists and pathologists, but also by those researching, developing and marketing new targeted anti-cancer therapies. Drug manufacturers now operate in a very different world than just a decade ago, and a solid CDx launch strategy is of vital importance. Such a strategy must ideally incorporate all the new advances, anticipate the upcoming revolutions, and ensure being a prescription driver rather than a barrier by navigating the ever-increasing complexity. Multiple markers tested in sequence First Next Generation Sequencing panels Exome sequencing, genome sequencing Proteomics Routine genome profiling of every patient s tumour First CDx tests. Single markers Truly personalised and individualised treatment Cancer conquered at the molecular level? Multiplex testing; simultaneous testing of several markers Significant uptake of large pan-cancer testing panels Routine ctdna testing Real-time tracking of novel mutations Fig 8: The past, present and future of Oncolology Companion Diagnostics) Cancer is complex, and an understanding of this complexity leads to opportunities: for doctors, for drug and diagnostics manufacturers and most importantly for patients.

HARNESSING COMPLEXITY: COMPANION DIAGNOSTICS IN ONCOLOGY 1 Ross JS et al. (2004) Targeted therapy in breast cancer: the HER-2/neu gene and protein. Mol Cell Proteomics 3: 379 398 2 Cobleigh M, Vogel C, Tripathy D et al. (1999) Multinational study of the efficacy and safety of humanized anti-her2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 17: 2639-2648 3 Mass R, Press M, Anderson S et al. (2001) Improved survival benefit from Herceptin (trastuzumab) and chemotherapy in patients selected by fluorescence in situ hybridization (abstract 18). Breast Cancer Res Treat 69: 213 4 Paik S et al. (2008) HER2 status and benefit from adjuvant trastuzumab in breast cancer. N Eng J Med 358: 1409-1411 5 Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N et al. (2009) Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361: 947-957 6 Zhu CQ, da Cunha SG, Ding K et al. (2008) Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol 26:4268-4275 7 Roche Molecular corporate website (http://molecular.roche.com) 8 Qiagen corporate website (https://www.qiagen.com) 9 U.S. Food and Drug Administration. 2013. Letter from Dept of Health & Human Services. [ONLINE] Available at: http://www.accessdata.fda.gov/cdrh_ docs/pdf12/p120019a.pdf. [Accessed 30 October 2015]. 10 U.S. Food and Drug Administration (http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id=14560) 11 U.S Food & Drug Administration. 2015. FDA expands approved use of Opdivo in advanced lung cancer. [ONLINE] Available at: http://www.fda.gov/ NewsEvents/Newsroom/PressAnnouncements/ucm466413.htm. [Accessed 17 February 2017]. 12 Genomeweb. 2016. FDA Expands Indication for Dako s PD-L1 Complementary Diagnostic. [ONLINE] Available at: https://www.genomeweb.com/ molecular-diagnostics/fda-expands-indication-dakos-pd-l1-complementary-diagnostic. [Accessed 17 February 2017]. 13 Agilent Technologies. 2016. Agilent Technologies Receives Expanded FDA Approval for Use of Dako PD-L1 IHC 22C3 pharmdx Companion Diagnostic in Non-Small Cell Lung Cancer (NSCLC). [ONLINE] Available at: http://www.agilent.com/about/newsroom/presrel/2016/24oct-ca16033. html. [Accessed 17 February 2017]. 14 Ipsos Healthcare Oncology Monitor, US, 2016 [research conducted online, Jan Dec 2016] Contact To find out more, or to discuss your specific research needs, don t hesitate to contact our Molecular Diagnostics team: (Global) Pieter De Richter Head of Molecular Diagnostics Portfolio Tel: +601 2290 0315 Email: Pieter.DeRichter@ipsos.com (US) Ayse Levent Vice President, Global MDx Monitors Tel: +1 646 313 7725 Email: Ayse.Levent@ipsos.com (UK) Dr Christophe Homer Associate Director, Global MDx Monitors Tel: +44 (0) 20 3059 4672 Email: Christophe.Homer@ipsos.com 19

Ipsos Healthcare About the Ipsos Oncology Center of Excellence With over 100 people dedicated to oncology worldwide and even more accredited through our Oncology Accreditation Program Ipsos has the fluency in oncology to meet our clients every need. Our Oncology Center of Excellence includes a comprehensive suite of custom capabilities alongside market-leading syndicated services such as the Global Oncology Monitor, the Oncology MDx Monitor and the Oncology Lab Mapping Study. Our key differentiator is our ability to integrate all oncology solutions to offer in-depth insights that effect tangible change. With unrivalled strength and presence in both developed and emerging markets, we always deliver global expertise with local market knowledge. www.ipsoshealthcare.com