Chapter 10
Creating Agility Through Data and Analytics

Introduction

It is difficult not to get caught up in the statistics associated with big data. An estimated 2.5 billion bytes of data are collected daily by a growing cadre of sensor-based technologies, from mobile phones to the Internet of Things—from connected “smart” thermostats in homes to desktop computers [1]. Consumers worldwide used more than six billion connected “things” in 2016 [2]. As a result, analysts predict the digital universe, defined as the data created or copied yearly, will grow to 44 zettabytes, or 44 trillion gigabytes, by 2020 [3].

That staggering amount of data has the potential to disrupt a number of different businesses, including traditional biopharmas. This phenomenon has already altered the retail, banking, and transportation industries in fundamental ways, as organizations such as Amazon and Apple mine customer-generated information for insights about buying habits and behaviors in order to offer customized buying experiences [4]. Change in healthcare has not been quite as rapid. Appropriate concerns about patient privacy and a highly regulated product development environment have made it more difficult for biopharma companies to extract measurable value via the large-scale integration and analysis of data.

Yet data, and the analytics platforms that are critical to make sense of the wide variety of relevant data sets, have the potential to address the biggest challenges in healthcare. As noted in prior chapters, unsustainable cost inflation and budgetary pressures have fueled a shift in drug reimbursement from fee-for-service to paying-for-outcomes. At the same time, we have witnessed an explosion in the volume, variety, and velocity of health data. These data include electronic health record data, payers' claims data, gene sequencing data, real-time data generated by mobile technologies, and patient-reported data on social media sites such as Facebook and Twitter and from patient communities and advocacy organizations (e.g., PatientsLikeMe).

What if biopharmas could harness these data to identify the small percentage of individuals most likely to consume a disproportionate amount of healthcare resources and design appropriate therapies and behavioral interventions to keep them healthy? What if biopharmas could integrate these data into their R & D programs to identify better drug targets and further enhance the efficiency of clinical trials? What if new data types could inform commercial activities to better demonstrate the value patient, payer, and provider stakeholders say they want? Put more simply, what if biopharma companies could combine these different streams of data to see the “big picture”?

Such questions set up a compelling vision, in which the aggregation and utilization of real-world, real-time data harvested from diverse sources has the potential to transform every aspect of the biopharma value chain. Some of this is already happening. However, biopharmas are only at the beginning of understanding how best to use data and which kinds carry the most currency with stakeholders such as payers.

It is also true that drugmakers will only be able to extract value from data—estimated by some analysts to be greater than $1 billion across a medicine's life cycle—if they can transition to an analytics-driven culture [5]. Making that leap requires that drugmakers overcome significant cultural, technological, and, in some cases, regulatory barriers to create new business practices that can replace—or coexist with—complicated processes that developed in a pre-digital age. As Charles Hansen famously put it, “All the problems with digital are analog problems” [6].

This chapter reviews the opportunities and challenges represented by the current explosion in data and analytics technologies. It also addresses the multiple forces that require biopharma companies to have access to data analytics expertise. If, in the future, intellectual property is tied less to an actual product than the real-world evidence that product generates—and the algorithms used to uncover it—it will behoove biopharma companies to develop robust systems that allow them to shorten the cycle time between data generation and the creation of business insights.

Multiple Forces Converge to Create Data and Analytics Opportunities

If biopharmas and their collaborators want to take advantage of today's data rich environment, they cannot just be excellent data aggregators. They must also be excellent data analyzers. Advances in a host of enabling tools, from cloud-based storage to natural language processing and machine learning, have resulted in an unprecedented opportunity to analyze and link disparate types of structured and unstructured health data, both big and small. Indeed, four different forces have recently converged to make data analysis a core capability within biopharmas (Figure 10-1). These four forces are:

  • Changing customer expectations
  • Changing speed of data generation
  • Changing reimbursement models
  • Changing biopharma business models

Figure 10-1 Multiple forces require biopharmas to become analytics experts

As discussed in Chapters 5 and 8, the traditional customers of biopharma products, physicians, have less voice in the current climate than a decade ago, while payers and patients have increasing power—and increasing expectations—due to economic shifts that require better population management as well as patient cost-sharing for medicines. In this environment, these groups of super-consumers want more data, not simply about product safety and efficacy but also about outcomes. The democratization of information and the growing importance of patient centricity mean that patients are not only informed about treatment options but are important influencers in the prescribing decision, especially when side-effects might adversely affect quality of life.

The changing speed of data generation is another important factor. As the wearable device revolution outlined in Chapter 9 accelerates, clinical-grade health data obtained by passive sensors linked to digital apps is improving. Indeed, Eric Schadt, the founding director of the Icahn Institute for Genomics and Multiscale Biology at New York's Mount Sinai Health System, predicts that as these devices grow more powerful “accurate information about your health will exist more outside the health system than inside the health system” [7]. That is both an opportunity and a challenge for biopharma companies. In order to take full advantage of this ever-larger pool of data, drugmakers must be able to mine it rapidly to make different business decisions.

Similarly, changes in healthcare reimbursement also require biopharmas to become more familiar with data analytics. As reimbursement is increasingly based on cost effectiveness and unique real-world value, data demonstrating that effectiveness are an intrinsic part of not only a product's value proposition but the basis for novel payment models that share risk between drugmakers and payers.

These changing customer expectations come at a time when biopharma business models are already under stress. R & D productivity has suffered as companies attack scientifically more challenging therapeutic areas such as Alzheimer's disease or attempt to develop breakthrough innovations in disease areas well served by generics (e.g., cardiovascular disease). Meanwhile, as drug spending becomes a greater concern in nearly every major market, companies have more limited ability to make regular price increases on existing products.

The end result is that as growth via traditional innovation has slowed, biopharmas are in the process of embracing new business models that go beyond selling products to creating solutions based on real-world data. At their core, these new business models are rooted in the ability to use data and analytics to adapt quickly to the evolving market. This ability to adapt becomes even more important as new entrants such as infotech companies enter the health space with their own data-driven solutions.

Healthcare's Four Data Vectors: Volume, Velocity, Variety, and Veracity

Since their beginnings, biopharma companies have been data-driven organizations. They are already adept at using data from randomized clinical trials to develop novel products that are safe and efficacious. In the evolving commercial landscape, biopharmas are now trying to simultaneously collect data to do the following: better segment patient populations, more accurately target high-prescribing physicians, collect real-world proof of outcomes to satisfy public and private payers, and incorporate these real-world findings into their discovery and research and development activities.

As data have grown in importance, the number of terms to describe it have likewise ballooned (see the box, “Important Definitions”). Most of the time, health data still do not clear the bar of being truly “big data” in terms of the amount of bytes being aggregated. The advent of large genetic data sets and information collected by Internet of Things (IoT) devices means the landscape is shifting rapidly, however.

Health data can be classified along four different axes: volume, velocity, variety, and veracity (Figure 10-2) [10]. In addition to anonymized patient record data, imaging records, biometric sensor readings from wearables, genetic sequence information, and social media interactions have driven tremendous growth in the volume, velocity, and variety of health data now being generated. While each of these three parameters present challenges for biopharma companies, it is the third “V”, variety, which has been the most difficult to manage.

Figure depicting a circle with data written in its center. From here four arrows point at volume, velocity, variety, and veracity. Cloud-based storage is mentioned between volume and veracity, natural language processing, AI is mentioned between volume and velocity, artificial intelligence is written between velocity and variety, and optimization, governance, and anonymization is written between variety and veracity.

Figure 10-2 Enabling tools to leverage data's four vectors

Consider volume, first. While the explosion of healthcare data is a challenge, the obstacles biopharmas face are really no different from those other industries (e.g., retail or consumer) have confronted. This means analytics tools that have been well validated elsewhere can be adapted, or applied, to the biopharma space, while management teams build the organizational structures required for analytics-driven cultures to flourish. Given healthcare's time frames, biopharma companies also do not need to use analytics to make decisions based on data generated in microsecond time frames. As such, managing the velocity of data generation in healthcare is not as big a concern as it is in industries such as finance or banking.

Instead, it is the variety of data biopharma companies must now assimilate that is the primary hurdle to clear. It is also one of the biggest opportunities. While each data type, for instance electronic health record data or genetic data or lifestyle data, has tremendous value on its own, it is only by linking these disparate pieces of information together that biopharma companies can achieve a step change in business as usual. As David Shaywitz, chief medical officer of DNAnexus, a leading provider of enterprise platforms for translational informatics, notes, “it is from the collisions of data where true insights arise” [11].

But each of these different data types exist in different formats and, because of patient privacy, have been stripped of identifying information. That makes combining the data into a credible whole challenging. Biopharmas must account for unstructured information—from social media commentary, physicians' notes in electronic health records, environmental readouts, and patient journey descriptions in clinical trials—while linking it to more structured data from the electronic health record in ways that can lead to new kinds of insights. In addition, the challenge of assimilating disparate data is made greater because most are owned by different players in the health ecosystem. Thus, if biopharma companies want to fully transform their businesses via integrating real-world data with patient health records, they must either purchase the information or partner with other healthcare stakeholders to access what they do not already own. As discussed later in the chapter, the dynamic market for healthcare data—and how best to partner with payers and providers, two groups that do not necessarily trust biopharmas—is of growing importance as the changing commercial landscape evolves to require integrated data covering the totality of the patient experience.

In addition to the three attributes described above, there is a fourth data characteristic that is critical to consider when evaluating health data: veracity. As biopharmas increasingly use unstructured data in their analytics efforts, they must take particular care to make sure the data are as error-free as possible and credible. As discussed later in the chapter, data quality is a particular challenge because unstructured data can be highly variable and inaccurate, for instance, due to mistranslation of a doctor's handwriting [12].

Extracting Value from Data Requires New Tools

As healthcare transitions from paper records, prescriptions, and imaging films to digital technologies, the information needs to be collected and stored on a common platform. In this environment, cloud computing has become a key enabler of data management. It not only allows easy access to information regardless of where the data are generated but supports the storage of disparate types of data in a common location. That makes the bits and bytes easier to manipulate. In addition, the existence of secure clouds from third-party specialists such as DNAnexus or Medidata Solutions means biopharma companies do not have to spend the time and money investing in the creation and upkeep of large departments devoted to data management and storage. That lowers the barrier to entry for big data analytics, particularly for younger biopharma companies, allowing them to scale their activities more quickly.

Jeffrey Reid, Ph.D., Executive Director and Head of Genome Informations at the Regeneron Genetics Center, a division of Regeneron that is applying high throughput genomics to speed drug discovery and development, notes the company's data efforts would have had a much rockier growth curve in the absence of cloud computing. Just setting up the data storage center would have required a massive effort, from the purchasing of hardware to the creation of a dedicated facility with computer servers. “From a pure infrastructure standpoint, we were able to go from not having a center to establishing a top of the line capability in a time frame that would not have been possible without cloud computing,” he says [13].

In addition to cloud computing, other tools capable of analyzing structured and unstructured data are essential as biopharma's pool of data grows. There is now a pervasive belief that having more data is always better. In reality, having more data can be a mixed blessing. “All you have really done,” notes David Davidovic, founder of the consultancy pathForward and a former senior executive at Merck & Co., Genentech, and Roche, “is build a bigger and bigger haystack of data that organizations do not know what to do with” [14]. Indeed, according to the market research firm IDC, “only about 5% of the data that are currently captured worldwide are ever analyzed” [15].

Getting a return from data will require deeper analysis of the other 95 percent of the information. Here tools that are making a difference include the divide-and-process approach of parallel computing, software platforms such as Apache Hadoop, the use of probabilistic statistical models, and advances in artificial intelligence (AI), including cognitive computing and machine learning. Such tools move beyond human-based applications to more predictive solutions that can both manage the proliferation of data and quickly generate meaningful insights to biopharma decision makers at a relevant point in time [16]. (See the boxes, “Using Machine Learning to Move from Experience-Based to Evidence-Based Medicine: The Evolution of GNS Healthcare” and “Artificial Intelligence: The Potential to Make Drugs Smarter, Faster, and for Less.”)

The Analytics Continuum: From Descriptive to Prescriptive

If biopharmas are to fully utilize the power of data and analytics to transform their businesses, their capabilities need to evolve from being descriptive to prescriptive. In other words, companies need to shift from identifying what has happened (e.g., drug sales in a given market grew 20 percent in six months) to understanding what steps to take to make the event happen (e.g., by taking a more strategic approach to market the product selectively to certain top regional health systems, which currently have low awareness of the product, it will be possible to increase product sales by the desired target). Moving from the “what” to the “how” occurs along a four-step continuum, which is described in Figure 10-3.

Figure 10-3 The analytics continuum

The first step in the process is the simplest: with descriptive analytics, biopharma companies need to consolidate data sources, creating systems that consistently collect, aggregate, and distribute structured and unstructured data from both internal and external sources. Mining the data typically requires real-time dashboards or other visual tools. At this stage, it may also be useful to perform diagnostic analytics to understand, based on past performance, why something happened. A biopharma preparing to launch a new cardiovascular product, for instance, might look at its three most recent launches to assess sales force deployment, advertising spend, use of co-pay cards, and other factors to gain a data-driven understanding of what worked—and what did not—in past launches. That understanding would allow the company to refine and further optimize its current launch strategy.

As biopharmas' expertise with data grows, they will advance to using analytics to identify past patterns that are predictive of future events. Such predictive analytics are particularly valuable when making complex forecasts, for instance, tied to a product or business unit's sales and marketing, as they help drive actual business decisions and help manage risk. Research shows even a modest improvement in the accuracy of predictions can result in significant savings or increased revenues. As Eric Siegel recounts in Predictive Analytics: The Power to Predict Who Will Click, Buy, Lie, or Die, one insurance company used predictive analytics to reduce its loss ratio, defined as the total amount it paid in claims divided by the total amount of premiums collected, by a half percentage point. The net effect of this small improvement was financially significant, worth around $50 million annually [21].

According to the technology research firm Gartner, only about 13 percent of businesses across all industries currently use predictive analytics. Fewer still, just 3 percent, utilize the most valuable form of analytics—prescriptive analytics [22]. Prescriptive analytics are designed to help answer specific questions, for instance, what combination of interventions, including drugs and behavioral change, could keep a patient with a history of heart disease, diabetes, and poorly controlled cholesterol healthy and out of the hospital? Having access to such data would obviously empower biopharma companies to make more informed investment decisions about services that might be wrapped around a product, or additional data to collect in the development phase to showcase value to stakeholders such as payers and health systems.

A cross-industry analysis by Forbes Insights suggests that when it comes to data capabilities, companies developing technology and consumer products have the most mature data and analytics capabilities, while biopharmaceuticals companies score close to the bottom, roughly on par with healthcare organizations and governments [23]. There are a number of reasons for this lower level of maturity. First, appropriately managing and manipulating health data requires access to a portfolio of analytics tools and skill sets that are different from the business intelligence tools biopharmas have traditionally employed. In addition, the highly fragmented healthcare market means considerable work is required to aggregate high-value data from different payers and providers, as well as patients and caregivers, to establish a complete picture of health needs and potential trends. Finally, healthcare is highly regulated and patient privacy remains a critical part of the social contract between patients and other stakeholders. How best to use algorithms to target high risk-patients while maintaining appropriate standards for sensitive, personally identifiable information is an issue yet to be adequately resolved [24].

Data Analytics Across the Biopharma Value Chain

For data to actually transform the biopharma value chain, companies will need to build capabilities or partner to access a portfolio of different tools and technologies. Indeed, one of the challenges in creating an analytics-driven organization is that no one single technology or process provides all the analytics capabilities required to answer all the questions that arise across the biopharma value chain, from how to discover the most effective biological targets, to how to develop them efficiently, to how to optimize product sales (Figure 10-4). As discussed later in the chapter, the possibility of siloed information or expertise makes it imperative that companies establish governance structures that promote appropriate sharing and integration of data.

Tabular representation of analytics across the biopharma value chain. The first column accounts for stage of value chain that includes discovery, development, supply chain, sales and marketing, and life cycle management. The second and third columns account for the corresponding role of analytics and example.

Figure 10-4 Analytics across the biopharma value chain (selected examples)

Improving Research and Discovery

Data analytics has perhaps the biggest potential to enable a new drug discovery and development paradigm, though currently the return on investment at this step of the value chain is hardest to measure. The advent of new genomic technologies has resulted in significant reductions in the cost of genome sequencing. According to the National Human Genome Research Institute, which collects data from the genome sequencing groups it funds, in mid-2015, it cost around $4,000 to create a high-quality draft whole human genome sequence. By late 2015, the price tag was dramatically lower—just $1,500; and if only the protein coding regions were desired—what scientists call the exome—the price tag was lower still, under $1,000 per genome [25]. As sequencing technology at Illumina and other biotechs develops, costs are anticipated to drop further, potentially under $100. As sequencing costs fall, biopharmas are able to rethink how they identify and validate drug targets, leveraging big sets of genetic data to find interesting but rare biological signals that could be the basis for novel medicines.

This approach grows even more powerful if biopharma companies are able to link unstructured phenotypic data, for instance, from patient electronic health records, to genetic data. Combining the genetic data with unstructured historical data via powerful analytics, biopharma companies can uncover not just previously hidden biomarkers, but relate their molecular research to outcomes that previously were not recognized.

Tapping that opportunity is one of the reasons Regeneron formed its Regeneron Genetics Center (RGC) and partnered with Geisinger Health System to sequence the genomes of hundreds of thousands of Geisinger patients [26]. In March 2016, the partners published their first peer-reviewed paper, using sequence data linked to de-identified longitudinal health records to identify a specific genetic mutation that results in a significantly reduced risk of coronary artery disease [27]. The findings corroborate ongoing R & D efforts at Regeneron from animal studies, as well as biochemical and early human genetic experiments, about the novel target's importance in improving clinical outcomes. “This new evidence gives us a lot of confidence about our development strategy and the ability to identify game-changing opportunities,” says Aris Baras, M.D., Head of the RGC [28].

In addition to Regeneron, start-ups such as Berg Health, TwoXAR and Sema4 are on their own, and in partnership with academic groups, bigger biopharmas, and infotechs, using cognitive computing and other big data tools to develop novel drugs for complex disease states [29–31].

Cancer has been one of the areas where these efforts to transform biopharma discovery have been most obvious; that is partly because the same trends driving convergence and precision medicine, topics outlined in Chapters 1 and 4, respectively, reinforce the demand for sophisticated analytics. As outlined in Chapter 1, IBM, via its Watson Health division, is making a clear bid to use cognitive computing to identify and personalize oncology therapies [32].

Going forward, this intersection of genetics and what has historically been health IT will merge and further drive the acceleration of efficient drug R & D. Critically, the failure rate for drugs in late-stage trials is still too high in a world of constrained resources. According to a team of researchers at Sagient Research Systems and the Biotechnology Innovation Organization (BIO), around 40 percent of all drugs fail in phase III [33]. Since the cost of R & D increases sharply from one phase to the next, such late-stage failures represent a very inefficient use of R & D capital. Because of historical pricing flexibility, biopharmas have not had the pressure to focus on R & D efficiency, but that situation is changing rapidly. In the future, biopharma companies will need to better use enabling tools such as artificial intelligence and causal machine learning to improve this portion of the value chain [34].

Improving Clinical Trials

There is no question the current clinical trials paradigm, defined as sequential phase I, II, and III studies punctuated by months of analyses and planning, is outdated. In sharp contrast to the technology and consumer sectors, where big data collected in real time routinely informs product formation, the basic framework for drug development has not changed since the 1960s [35]. Because of the high cost of late-stage drug failures, however, biopharmas are beginning to embrace analytics-driven clinical trial designs to increase the efficiency and success rates of their development programs. These so-called adaptive designs rely on Bayesian algorithms to enable preplanned adjustments to clinical trials to refine hypotheses and reallocate R & D dollars in real time based on clinical data [36].

In addition, as described in Chapter 9, the marriage of clinical data and social media (e.g., Twitter, Facebook, and YouTube, and especially online patient communities) creates a new opportunity to develop clinical descriptions of disease that can inform and accelerate clinical trial enrollment, while –omic data (e.g., genomic, proteomic, microbiomic, etc.) enable the identification of patients likely to respond. Similarly, the incorporation of digital technologies into trial designs allows the detection of early safety and efficacy signals. Indeed, data streams from wearables and sensors allow real-time feedback, creating an opportunity for continuous learning in clinical trials via analytics.

Biopharma companies are only now beginning to identify how to leverage analytical tools such as machine learning in this regard. Many of the first efforts are clustered in neurological disease areas where measuring product efficacy and disease progression have historically depended on subjective survey data. Teva Pharmaceutical Industries, for instance, teamed up with Intel in September 2016 to incorporate data from wearable devices in a phase II trial monitoring disease progression in Huntington's disease to generate objective scores measuring the severity of motor symptoms [37].

Creating the Agile Supply Chain

As cost pressures mount and competition from both start-ups and new entrants increases, larger biopharmas are increasingly focused on optimizing their operational performance, especially the activities tied to manufacturing and supply chain. Advanced analytics has a key role to play here. Beyond improving manufacturing efficiencies, analytics can help companies do a better job of creating real-time forecasts, reducing the need to carry costly excess inventory, as well as creating new drug distribution platforms.

A 2014 study shows that while most biopharmas amass significant amounts of historical manufacturing and supply-chain related data, the collected statistics are siloed across different parts of the organization and the available tools are too limiting to handle biopharma's complex manufacturing processes [38]. As streamlining business processes becomes more important in today's tougher climate, however, biopharma companies will look outside the industry to lessons learned in the retail and consumer technology sectors. Analytics-enabled flexible supply chains like the kind Apple employs to create its iPhones will become the norm. Steps that can optimize the hand-offs between the wholesaler, the retail pharmacy, and the patient encourage care coordination and adherence, which can ultimately promote a drug's value

To that end, GlaxoSmithKline in 2014 teamed up with Cambridge University's Institute for Manufacturing to build a more efficient end-to-end supply chain that brings together equipment manufacturers, regulators, knowledge transfer networks, and healthcare providers [39]. Three years earlier, the biopharmaceutical company also partnered with what, at the time, was a non-obvious player: the Formula One race car manufacturer McLaren. On the racetrack, every second counts, so pit crews use sophisticated data analytics to reduce the time required for tire changes and other repairs. GlaxoSmithKline wanted to adapt such methodology to its own processes, particularly in its consumer health business, where small-batch manufacturing leads to not just lost productivity but lower margins. After working with McLaren for about a year, GlaxoSmithKline reduced downtime in one particular manufacturing plant by 60 percent. Since 2011, the two organizations have worked to scale-up changes and have expanded their efforts to make R & D more efficient via the use of sophisticated sensors in clinical trials [40].

Using Analytics to Rethink Commercial Activities

The shift from fee for service to fee for value and the growth of digital technologies have made what was once one of the most straightforward parts of the biopharma value chain—sales and marketing and life cycle management—one of the most complicated. Indeed, estimates suggest that approximately two-thirds of new drugs fail to meet prelaunch sales expectations in their first year on the market and continue to underperform for the next two years [41].

As noted in Chapters 7 and 8, one reason so many recent drug launches have been suboptimal is the need to have, at launch, data showing real-world utility. Absent such information, increasingly skeptical payers and providers make medicines harder to obtain, either by putting them on expensive formulary tiers or by excluding them altogether [42]. (See the box, “Real-World Data—and Analytics—Play an Increasingly Important Role in Establishing Product Value.”)

Beyond real-world evidence analysis, other commercial activities that will alter as a result of data analytics include strategic payer engagement, a topic discussed in Chapter 8, and the channel and geographic optimization of a biopharma's sales force via multichannel digital tools. Such analytics capabilities better enable drugmakers to interact with the right physicians, key opinion leaders, and health systems based on a therapy's clinical profile. As it becomes increasingly difficult to engage with already busy physicians, the ability to customize the message to the right prescribers is essential. On average, high-prescribing physicians are contacted by drugmakers nearly 3,000 times a year. But many of those interactions are overly general and come as physicians are struggling to deliver higher quality care in shorter appointment times [46].

While there is a major opportunity to make the experience for the provider more tailored and personal, the potential to optimize the patient experience could be even greater. Luca Foschini, Chief Data Scientist at Evidation Health, which develops predictive analytics tools for a variety of healthcare stakeholders, believes the same arc that has enabled customer-centricity in the online advertising and retail spaces will also fundamentally change the patient experience. “Personalization was completely absent 10 years ago and poorly understood five years ago,” he says. “Now it is making a huge difference” [47]. Indeed, as organizations such as Netflix and Amazon have grown more data-savvy, they have embraced predictive analytics to mine a user's viewing or reading habits to make recommendations on future movies or books, further customizing the experience and changing consumer expectations [48].

As biopharmas develop their analytics capabilities, one can envision them taking similar strides to customize the patient experience either directly, where regulations permit, or through collaboration with providers. They might use wearable data to provide tailored interactions to patients based on their health status, for instance, to promote medication adherence or healthy behaviors such as better sleep hygiene. Data analytics could also be a valuable tool for connecting more directly with patients on aspects of disease education. Mining metadata from social media (e.g., Twitter, Facebook, and YouTube), biopharmas could begin to develop clinical descriptions of disease that are consistent with how patients themselves discuss their symptoms. This terminology can then be used to develop more meaningful education-based and patient-based portals.

Biopharmas interested in these new approaches must tread carefully, however. Guidance on how biopharmas can interact with patients safely and legally are still evolving, and there is clear evidence that patients want brand-agnostic information, not materials linked to a specific therapy. Indeed, success will depend partly on establishing a new social contract that makes patients more willing to share their personal data with non-provider stakeholders in the health space [49].

Future Uses

As drug companies fully integrate external and internal data to answer stakeholders' needs and gather product evidence about value, they have an opportunity to create new types of data-rich partnerships that are less transactional (selling pills) and more relational (engaging around health outcomes). (See the box, “The Dynamic Data Market.”) These include new risk-sharing models, described in Chapters 7 and 8, as well as beyond-the-pill service models where the intellectual property is not a therapeutic but an algorithm.

Consider that biopharmas can partner with payers to combine the latter's claims and authorization data with its proprietary clinical trial data and develop algorithms to forecast the monetary impact associated with using a novel but relatively untested product versus the potential benefits. The biopharma could devise a pricing contract that preserves pricing flexibility but limits the potential monetary risk to the payer if utilization exceeds the forecast. This kind of analytics-based outcomes payment can improve the biopharma's productivity, accelerating the collection of valuable outcomes data while simultaneously generating revenue. An added but important bonus: by sharing data in this manner, the biopharma strengthens its relationship with a critical customer.

Companies are already edging into this arena. For example, Vifor Fresenius Medical Care Renal Pharma, a joint venture between Vifor Pharma and Fresenius Medical Care, is developing an outcomes-based service to predict, and ultimately prevent, debilitating anemia in patients with chronic kidney disease. The service hinges on structured data collected from laboratory reports. As enabling technologies such as cloud computing and machine learning evolve, however, future algorithms will also take advantage of environmental and personal data as well [50].

Data and Analytics Challenges

As biopharmas gain expertise with data analytics, there are a number of challenges they must overcome (Figure 10-6). These challenges can be grouped into the following categories:

  • Data integration: How can companies abolish data silos and integrate external and internal data across the organization?
  • Data quality: With the huge increase in data volume, how can companies prevent inconsistencies that skew the analysis?
  • Data privacy: How can companies remain compliant with privacy, regulatory, and security issues?
  • Pace of change: With the field advancing so quickly, where are the biggest returns and how should biopharmas invest?
  • Organizational and cultural barriers: How can incentives be created that promote a data-first business orientation when return on investment is still unclear?

Figure 10-6 Challenges blocking wider usage of data and analytics

Data Integration

The first two challenges, data integration and data quality, are largely technological hurdles that companies have started, albeit slowly, to address. Because of the fragmented nature of health data generation—via wearables, medical records, lab tests, and payer claims—one of the first steps is gaining access to the relevant data. As biopharmas invest more heavily in real-world evidence collection post-launch, they will generate some of these data themselves. Other types of data, for instance, anonymized electronic health record information or claims data, will come via partnerships with providers or payers. As noted, biopharmas are already active in this arena. Still other data, for example, air quality forecasts or consumer search patterns, can be purchased from third-party sources, including infotech players.

In addition to obtaining access to the needed data, biopharmas must create a common platform, whether via a third-party cloud or an internally built warehouse, to promote those all-important collisions of data (Figure 10-7). That common location encourages users to make connections between different types of data that were historically stored in discrete so-called “data lakes” inside or outside the organization. By integrating those different flows of data, the objective is to turn individual lakes into a larger data ocean. At even the most sophisticated biopharmas, however, the current level of in-house analytics capabilities makes this prospect more an aspirational goal than a reality.

Figure depicting six rightward arrowheads corresponding to biopharma (preclinical research, clinical trial data, biomarker analysis), providers (health record data, hospital admissions, etc.), payers (claims data from pharmacy, hospital, and laboratory), pharmacy (product sales, script volumes, medication adherence), laboratories (diagnostic volume, biomarker assessment), and patients (social media commentary, sensor data, quantified self/fitness tracking).

Figure 10-7 Data remain siloed within healthcare's diverse ecosystem

To encourage data sharing, especially across large biopharma organizations, management teams will need to develop systems that consistently structure data in ways that preserve contextual relationships, even when the information has been de-identified due to privacy regulations. This is particularly important for unstructured data, as well as non-health data that has an impact on patient wellness (e.g., environmental data related to air quality for patients with asthma or chronic obstructive pulmonary disease). As such, strict data governance protocols, including using master data management techniques that resolve inconsistencies in how data are defined, are important to establish early on. Indeed, researchers at the MIT Sloan Center for Information Systems Research recommend that bigger companies consider designating a “data dictator” to establish common definitions and data management protocols for the safe and appropriate handling of data [54].

Data Quality

For data to be useable and generate new insights, it has to be of high quality. This is not exactly a new problem for biopharma data teams, but it has been made worse by the volume of data being generated and the tendency for it to be stored in silos. The problem is, if the “small data” are not trustworthy, having a bigger pool does not necessarily help. It just increases the risk that biopharmas draw erroneous conclusions based on their analyses. Moreover, if biopharmas are to avoid the garbage in, garbage out phenomenon, they must actively avoid what researchers at Northeastern University and Harvard University term “big data hubris,” in which “big data are a substitute for, rather than a supplement to, traditional collection and analysis” [55]. (See the box, “Google Flu Trends' Unanticipated Big Data Lesson.”)

Data Privacy

In many markets, privacy regulations require healthcare organizations to remove patient identifying information before the information is shared. That can make it more difficult to conduct the predictive and prescriptive analytics most likely to offer the biggest return on investment. For instance, a biopharma company may want to demonstrate the value of a new cardiovascular medicine that might compete with older, cheaper generics. It could use predictive and prescriptive analytics to identify the 20 percent of the patient population that is at greatest risk of a costly adverse event and begin to conduct clinical and observational trials to showcase the value. But to build a complete picture, the biopharma will likely want to integrate a range of different data, for instance, information transmitted from digitally enabled scales or activity trackers. Without established processes for data governance and integration, it could be difficult to combine the data in meaningful ways that give a complete and actionable picture.

There are also emerging compliance risks for biopharma companies. As data are combined from different organizations, there is still the risk of re-identification, particularly if health data are combined with governmental information such as US voter registration details. To limit the risks, data management techniques that include the aggregation of smaller sample sizes are helpful [57].

But biopharmas will also have to review their own cybersecurity plans to make sure adequate safeguards exist for the highly sensitive patient data stored within their organizations or in the cloud. Data generated from IoT-enabled devices such as smart asthma or glucose monitors could be particularly vulnerable to hacks by cybercriminals who might use details to commit fraud [58]. Indeed, even if no hack actually occurs, the threat of a potential cybercrime can damage a company's reputation and its product revenues. That is what happened to Hospira in 2015, when the US Food and Drug Administration issued a warning about software vulnerabilities associated with the manufacturer's Symbiq drug pumps. Although Hospira initially worked on software updates to prevent remote hacking, it ultimately decided to remove the product from the market [59].

Though many biopharmas clearly understand the potential business risks cyber threats pose, a 2015 survey by EY found significant gaps in capabilities. Interestingly, 65 percent of life sciences organizations surveyed revealed a significant cyber incident that was not otherwise identified by their security team; a similar percentage, 61 percent, also noted they do not have a dedicated cybersecurity role to focus on threats tied to emerging technologies. Closing this capability gap will be important as biopharmas rely more heavily on big data [60].

Pace of Change

In addition to the challenges described above, biopharmas developing data analytics capabilities face a fourth major challenge: the pace of change. It may be surprising, but just five years ago, electronic health records were not widely implemented across health systems, and wearable usage was limited to early adopters. Since then, there have been dramatic improvements in enabling technologies that ease data access and integration, and specific analytics tools required to make sense of the information are now available. The question for biopharma companies in this dynamic environment is, how do you invest in data analytics in ways that are flexible and scalable?

This is not a trivial question. Typically, these kinds of information technology investments take months, if not years, to implement, along with large amounts of capital. Given the velocity with which technology standards and platforms change, and the fact that biopharma business models themselves are evolving because of macroeconomic forces, there is a real risk that, by the time a biopharma creates an internal analytics engine, it will no longer be fit for purpose. That is a potential problem when revenue growth at many of the biggest biopharmas in the industry is already slowing.

Organizational and Cultural Barriers

It is relatively easy to create a narrative that draws a direct line between improved data analytics and better market performance. Unfortunately, the data proving the value of data largely do not yet exist; one exception, perhaps, is the use of data analytics to optimize marketing to key providers. Given the potential significant upfront costs associated with creating analytics expertise, this lack of clarity about return on investment creates a further disincentive to accelerate investment.

In addition, there are other cultural and organizational barriers that might limit willingness to invest in analytics. Big data analytics is not a core capability for most biopharma organizations. Traditionally, data experts sat in a backroom buying and manipulating databases. Data was not seen as a strategic imperative that will accelerate work across all aspects of the biopharma value chain. This siloed thinking makes it more difficult to integrate different kinds of data across the biopharma organization and, thus, reap full value for the investment.

Biopharma firms also need new kinds of expertise, particularly data scientists able to extract meaning from various types of structured and unstructured data. Not only do such individuals understand the limitations of different types of data, but they are capable of quickly building predictive models to reflect real-time changes, for example, in product sales or clinical trial recruitment.

Individuals with these capabilities are a rare commodity: the strategic consulting firm McKinsey predicts a global excess demand of 1.5 million data scientists [61]. And since many data scientists come from mathematics or engineering backgrounds, these executives will also need healthcare-specific training [62].

Building an Analytics-First Organization: Cultural not Technical Hurdles

A 2015 survey of nearly 3,000 managers in different industries in MIT Sloan Management Review found that the chief obstacle to extracting value from analytics was not technological in nature, for example, data management or complex modeling skills. Instead, it was translating analytics into actual behavior change, or moving from big data to bigger insights [63]. So what should biopharma companies do to derive full value from their analytics efforts and create agility? There are three critical steps:

  1. Make analytics a strategic imperative
  2. Encourage the right behaviors
  3. Partner where possible for flexibility

Analytics as a Strategic Imperative

Inexpensive computing power, the cloud, and increasingly robust algorithms powered by artificial intelligence are the primary technological drivers behind the current data analytics opportunity. However, these technology advances should be viewed as necessary, but not sufficient, for success. The overarching goal is to institutionalize a set of behaviors that promote using analytics as a core competency to generate better business insights. Such sweeping behavior change will only come about when biopharma's senior leadership views analytics as a strategic imperative.

That means having a top-level executive committed to analytics initiatives, who is capable of advocating for resources and elevating the subject to the board as appropriate. This person could be a chief information officer (CIO) or a chief data officer (CDO). The title is less important than the ability to have someone responsible at a business-wide level for articulating how analytics will support the overall business strategy. “We see many organizations that have spun up initiatives and are spending a lot of money, (but) do not necessarily have a clear point of view on how value will be delivered,” notes Chris Mazzei, chief analytics officer for EY [64]. Indeed, a CIO or CDO can help connect the dots between interesting ongoing pilots so the companies can reduce duplication and translate successful initiatives to other groups in the organization.

Changing Behaviors

One of the key jobs for management is to create the incentives that promote the appropriate sharing of data and analytics tools across the organization. This is both a people management issue as well as a technological issue. For larger companies, one way to accelerate the process is to prioritize the creation of a center of excellence focused on developing certain crucial analytics processes. This senior management team would be responsible for doing the following: developing protocols for data governance and aggregation; centralizing methods, tools, and models so they can be shared easily across the organization; creating shared metrics for success; and promoting new innovations. By focusing on these kinds of activities, senior management can drive behavior change, promoting an environment where the results of prior analyses are factored into new analytics projects. Importantly, biopharma companies that use this approach spend less time searching for the right data and tools and more time performing analyses to drive important business decisions.

For this approach to succeed, however, the activities of the center of excellence must be closely integrated with the actual business. Having a team of data scientists produce multiple product launch scenarios that are then handed off to a biopharma brand unit to assess and implement is less likely to succeed than if the sales and marketing people interact with the analysts as a single team to develop a precision launch plan. Only by working together can the analysts and the business organization build credibility with each other and establish clear alignment between the end analyses and the business unit's priority—in this example, a successful launch. Indeed, absent a strong working relationship between business leaders and data scientists, there is a real risk that biopharma decision makers revert to business as usual, making decisions that are not driven by analytics and evidence but rooted in their preconceived notions and historical biases.

Partner for Flexibility

Bigger biopharmas face the option of building their own in-house capabilities either organically or via acquisitions, while smaller, earlier-stage biotechs, because of capital constraints, will almost certainly need to partner to access the data, the platform-enabling analytics tools, and the human expertise. However, just because bigger companies have the wherewithal to “go it alone” does not mean they should. Building a robust analytics engine requires significant up-front investments in time and money to create the appropriate infrastructure and capabilities. Because the technology is changing so fast, there is a real risk of spending hundreds of millions of dollars to create a platform that is unusable or out of date given the changing healthcare market.

To maintain flexibility, companies should consider whether an “as a service” model can be applied to various aspects of data analytics (Figure 10-8). Many biopharmas already outsource certain elements of their business, partnering with third parties around manufacturing or aspects of research like assay development. Why not analytics as well?

Figure depicting building the agile, analytics-enabled biopharma organization where the upper panel corresponds to functions that include discovery, research and development, regulatory, supply chain, sales and marketing, and finance/operations. A downward arrow from here points at analytics as a service that includes common data platform, shared data sources, cross-functional talent, shared analytics network, change management, and external partnerships.

Figure 10-8 Building the agile, analytics-enabled biopharma organization

Companies can partner with various third parties to get access to the capabilities that are most valuable based on their maturity and life cycle. A pre-commercial biotech in late-stage clinical trials could begin working with a commercial analytics specialist to develop analytics to optimize a pending product launch; a commercial biotech with just a handful of products might partner with an analytics provider to develop more customized pricing agreements with key health systems. Even larger biopharmas might be interested in working with bigger analytics players to integrate patient-centric data into their clinical trial design given changing regulations about when and how such data should be used. Simply put, by taking an analytics-as-a-service approach, biopharmas can scale-up—or scale-down—their use of analytics assets based on their changing needs. They can also limit their risks by contracting with a third party that can help generate the analytical insights.

Many of the biggest biopharmas will examine the analytics as a service option and decide that data-driven insights are such a core part of their business that they need to “own” these capabilities. Indeed, companies that are serious about moving beyond the pill may find that an essential component of their intellectual property is an algorithm designed to maximize product sales following launch. In this instance, companies will want to retain full control of the algorithm, and that means keeping the capabilities in house.

Even in this situation, given the variety of data that play a role in health, biopharma companies will need to establish partnerships with third parties. At a minimum, this means reaching outside the organization to access different types of relevant data to combine it with internally available data. But it also may mean sharing data with partners or suppliers to create new ways of doing business, for example, working with a payer to create an outcomes-based pricing model for a new but expensive drug. As the need to pursue such partnerships grows, companies will want to structure these agreements so that they retain control of the processes resulting in their data-driven insights.

Conclusion

Data and analytics will be a core competency for biopharmas going forward. For companies that are only now building these capabilities, the good news is that it is not too late. Those companies that grasp how data and analytics can improve fundamental aspects of the biopharma business, and, as a result, change their practices, will have a competitive advantage, outperforming the competition. However, to be successful, management teams must focus on how data and analytics enhance the value proposition of their companies' medicines. In doing so, they can actively avoid being edged out by infotechs that play an increasingly outsized role in health data generation via consumer-focused devices and software. As is true for so many other areas of biopharma drug development, from marketing to pricing to alliance building, context is everything.

Summary Points

  • Data, and the analytics that underpin them, have the potential to identify new solutions to some of healthcare's biggest challenges.
  • As business models put increasing primacy on customers' definitions of drug value, combining new types of data to generate outcomes-based insights is an essential skill for biopharmas.
  • Big data and analytics will alter every aspect of the biopharma value chain, especially commercial practices.
  • The return on investment for analytics capabilities is intuitive but, as yet, difficult to quantify, especially for earlier stages of drug development.
  • To build an analytics-first culture that takes full advantage of big data, biopharmas must access, or build, key capabilities, including tools to aggregate and analyze data.
  • As part of the process, senior biopharma leaders must remain focused on the questions that they want to answer, while avoiding data overload. The question of greatest importance is, “what is the business issue we really want to solve?”
  • To be successful, biopharma companies will need to take a portfolio approach, marrying specific types of big data and analytical tools depending on the disease area and competitive market dynamics.
..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset