HomeBlogTechnologyBeyond the Microscope: How Analytics is Transforming Biopharma

Beyond the Microscope: How Analytics is Transforming Biopharma

Biopharma data analytics: Top 3 Imperatives

Why Biopharma Data Analytics is Essential for Modern Drug Development

Biopharma data analytics has become a strategic necessity for leading the next wave of medical breakthroughs. The industry faces soaring drug development costs (over $2 billion per drug), complex regulations, and rising patient expectations for personalized medicine.

Key statistics highlight the urgency:

  • Market Growth: The life science analytics market is projected to reach $37.20 billion by 2030 (CAGR 13.6%).
  • Data Volume: Life sciences will soon account for 36% of all global data.
  • ROI Potential: Analytics can deliver 10-20% lower conversion costs and 10-15% better cost of quality.

While challenges are immense, the industry generates massive data volumes across the drug lifecycle. The opportunity to transform this data into life-saving insights has never been greater. Recent industry case studies show the potential: generative AI has identified preclinical candidates in as little as 18 months for about $2.6 million—a fundamental shift to predictive decision-making.

The question is no longer if companies should adopt analytics, but how quickly they can build a compliant, scalable foundation to gain a competitive advantage.

I’m Maria Chatzou Dunford, CEO and Co-founder of Lifebit. With over 15 years in computational biology and AI, I’ve focused on building solutions for genomics and biomedical data, helping pharma organizations implement biopharma data analytics in secure, federated environments.

Infographic showing the exponential growth of the biopharma analytics market from $15.27 billion in 2023 to $37.20 billion by 2030, alongside the increasing cost of drug development exceeding $2 billion per drug, and the massive volume of healthcare data representing 30% of global data generation with life sciences projected to reach 36% by 2025 - Biopharma data analytics infographic

The Strategic Imperative of Biopharma Data Analytics

The pharmaceutical industry is at a critical crossroads. On one hand, the potential for groundbreaking therapies has never been greater, driven by advances in genomics, cell therapies, and personalized medicine. On the other, the traditional R&D model is buckling under the pressure of spiraling drug development costs, high failure rates, and increasing regulatory scrutiny. This environment creates a clear and urgent imperative: harness data intelligently or be left behind. The shift is already happening, with around 80% of biopharma leaders planning to adopt AI and advanced analytics, recognizing it as a core competitive differentiator.

This paradigm shift is known as Biopharma 4.0, the next evolution of the industry that mirrors the principles of Industry 4.0. It involves the deep integration of AI, the Internet of Things (IoT), and connected data systems across the entire value chain—from the earliest stages of discovery to post-market surveillance. The financial impact is not theoretical; it is tangible and significant. Companies that successfully implement analytics are already reporting 10-20% lower conversion costs in manufacturing and a 10-15% improvement in the cost of quality. These are not just operational tweaks; they are direct boosts to EBITDA, freeing up capital and accelerating the entire R&D engine.

At the heart of this transformation is the drive toward personalized medicine. The blockbuster, one-size-fits-all drug model is giving way to highly targeted therapies designed for specific patient subpopulations. This requires the large-scale integration and analysis of incredibly diverse datasets: multi-omics data (genomics, proteomics), electronic health records (EHRs), clinical trial data, and real-world evidence (RWE). Without a sophisticated, scalable analytics foundation, the vision of personalized medicine remains unattainable.

To understand how intelligent data management drives these strategic outcomes, explore our insights on Data Intelligence.

Three converging trends are creating a perfect storm, accelerating the adoption of data analytics as a strategic necessity.

  • The Era of Personalization: The future of medicine is personal. The move from blockbuster drugs to precision therapies requires a profound understanding of disease at the individual level. This means analyzing vast patient datasets to stratify populations based on genetic markers, lifestyle factors, and clinical history. Biopharma data analytics is the engine that powers this stratification, enabling the design of targeted treatments and companion diagnostics that identify which patients will benefit from a specific therapy. This not only improves efficacy but also enhances safety and value, which is increasingly demanded by payers and regulators.
  • The Maturation of AI-Driven Discovery: Generative AI, deep learning, and natural language processing are no longer just academic concepts; they are practical tools revolutionizing drug discovery. AI algorithms can now predict how proteins will fold, design novel molecules with desired properties, and scan millions of scientific papers to uncover hidden connections between genes and diseases. The recent industry case studies—identifying a preclinical candidate in approximately 18 months for around $2.6 million—are not outliers but indicators of a fundamental shift. AI is de-risking the earliest, most uncertain stages of R&D, allowing scientists to focus their efforts on the most promising candidates.
  • The Rise of Biopharma 4.0: This paradigm extends beyond R&D to encompass the entire operational landscape. It involves creating ‘smart’ labs and factories where IoT sensors on equipment stream data in real time. It includes the development of advanced therapies like cell and gene therapies, which are inherently data-intensive and require meticulous tracking and real-time analytics for manufacturing, quality control, and supply chain logistics (chain of custody and identity). Biopharma 4.0 necessitates a connected data ecosystem, making data-intensive partnerships and patient-centric approaches not just possible, but essential for success.

Measuring Success: KPIs for Analytics Impact

The true value of biopharma data analytics is measured by its direct impact on critical business and scientific key performance indicators (KPIs). A successful analytics strategy is not an academic exercise; it drives measurable improvements across the organization:

  • Reduced batch failure rates: By applying multivariate analysis to real-time sensor data from bioreactors, predictive models can detect subtle deviations from the ‘golden batch’ profile, alerting operators to potential issues long before they lead to a costly failure.
  • Improved yields: Analytics helps optimize complex process parameters (like temperature, pH, and nutrient feed) in real time, pushing yields closer to their theoretical maximum and increasing throughput without additional capital expenditure.
  • Faster clinical trial recruitment: By analyzing real-world data and historical trial performance, analytics platforms can identify geographic hotspots with ideal patient populations and high-performing clinical sites, drastically cutting down recruitment times—often the biggest bottleneck in clinical development.
  • Better On-Time-In-Full (OTIF) delivery: Predictive analytics applied to supply chain data can improve demand forecasting, optimize inventory levels, and identify potential disruptions, building a more resilient and efficient supply chain.
  • Lower cost of quality: Proactively identifying and mitigating risks in manufacturing and supply chains prevents deviations, reduces the need for investigations, and lowers the overall financial burden of maintaining quality standards.
  • Shorter R&D cycle times: AI and analytics accelerate nearly every phase of development, from identifying targets and optimizing lead compounds to designing more efficient clinical trials and streamlining regulatory submissions.

These operational gains are not isolated wins. They translate directly to improved EBITDA, enhanced market competitiveness, and a faster path to delivering innovative medicines to patients. This proves that a robust analytics capability is a powerful strategic differentiator, not a mere cost center.

Understanding and leveraging real-world data proves crucial for many of these KPIs, particularly in clinical and post-market phases. Learn more about how we harness Real-World Data to drive these improvements.

Unlocking Value Across the Biopharma Lifecycle

Biopharma data analytics is not a siloed function; it is a transversal capability that creates profound value across the entire lifecycle. It transforms the deluge of data generated in labs, clinics, and factories from a logistical challenge into a strategic asset, providing actionable insights that accelerate timelines, de-risk development, and reduce costs.

Biopharma value chain with data analytics icons at each stage - Biopharma data analytics

Accelerating R&D and Discovery

In the high-risk, high-reward world of early-stage R&D, analytics transforms drug discovery from a process of serendipity and brute-force screening into a precision-guided, data-driven science. This is where analytics can have the most dramatic impact on the long-term pipeline.

  • Target Identification and Validation: Instead of relying on traditional methods, AI algorithms can now integrate and analyze population-scale genomic data (from sources like the UK Biobank or internal patient cohorts), proteomics, and transcriptomics to identify novel genetic variants or pathways strongly associated with a disease. Natural Language Processing (NLP) further enhances this by mining millions of scientific publications and patents to build evidence networks, helping scientists validate these targets and prioritize the most promising ones.
  • Multi-Omics Integration: Modern biology recognizes that disease is a complex, multi-layered process. Analytics platforms are essential for integrating different ‘omics’ data types—genomics (the DNA blueprint), transcriptomics (which genes are active), proteomics (the resulting proteins), and metabolomics (the metabolic processes). By creating a holistic, systems-biology view, researchers can understand disease mechanisms more deeply and identify more effective points for therapeutic intervention.
  • Predictive Toxicology and ADMET Profiling: A huge percentage of drugs fail in development due to unforeseen toxicity. In silico toxicology uses machine learning models trained on vast historical datasets of compound structures and their known effects to predict a new molecule’s ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) profile. This allows chemists to flag and design out potential liabilities—like liver toxicity or cardiac side effects—before a single physical experiment is run, saving millions of dollars and years of wasted effort.
  • Biomarker Discovery & Drug Repurposing: Analytics excels at finding complex patterns in clinical and omics data to identify biomarkers. These can be prognostic (predicting disease progression), predictive (predicting response to a specific drug), or pharmacodynamic (showing the drug is engaging its target). AI can also accelerate drug repurposing by systematically scanning for connections between existing drugs’ molecular profiles and the profiles of other diseases, offering a much faster, lower-risk path to new indications.

These advances are what enable breakthroughs like moving from target discovery to a preclinical candidate in just 18 months. To understand how these advances are reshaping the industry, explore our insights on AI in Drug Development.

Optimizing Clinical Development

Analytics brings much-needed efficiency, precision, and patient-centricity to clinical development, the most expensive and time-consuming phase of bringing a drug to market.

  • Intelligent Patient Stratification: By integrating genomic and clinical data, analytics can identify patient subgroups most likely to respond to a therapy. This allows for smaller, more targeted ‘basket’ or ‘umbrella’ trial designs that have a much higher probability of success. For example, in oncology, trials can be designed specifically for patients with a certain genetic mutation (e.g., KRAS G12C), leading to dramatic response rates and faster approvals.
  • Strategic Site Selection: A common cause of trial delays is poor site selection, leading to failed enrollment targets. Analytics platforms can mine real-world data, EHRs, and historical trial performance data to pinpoint clinical sites with the right infrastructure and access to the specific patient populations needed for a trial, ensuring faster and more complete recruitment.
  • Decentralized Clinical Trials (DCTs): Analytics is the backbone of DCTs. It enables the secure collection and analysis of vast streams of data from wearables (e.g., continuous glucose monitors, activity trackers) and ePROs (electronic Patient-Reported Outcomes). This provides a richer, more continuous, and more realistic picture of a drug’s efficacy and safety in a real-world setting compared to periodic, artificial clinic visits.
  • Synthetic Control Arms (SCAs): In many cases, especially in rare diseases or oncology, recruiting a placebo or standard-of-care control arm can be ethically challenging and slow. SCAs use curated, high-quality real-world data from sources like EHRs and disease registries to create a virtual, matched control group. This can accelerate trial timelines, reduce costs, and make trial participation more attractive for patients. Regulatory bodies like the FDA are increasingly open to well-designed studies incorporating SCAs.
  • Real-World Evidence (RWE): Beyond SCAs, RWE and Health Economics and Outcomes Research (HEOR) analytics are crucial for demonstrating a drug’s value to payers and securing market access and favorable pricing. This involves analyzing large-scale claims and EHR datasets to show how a drug performs in a broad, real-world population outside the confines of a controlled trial.

Learn more about how we’re changing clinical research through Real-World Data in Clinical Research.

Revolutionizing Manufacturing and Supply Chain

In manufacturing, analytics drives operational excellence, moving the industry from reactive problem-solving to proactive optimization and control.

  • Process Analytical Technology (PAT) and Multivariate Data Analysis (MVDA): PAT is an FDA-endorsed framework for designing, analyzing, and controlling manufacturing through timely measurements of critical quality and performance attributes. This is powered by MVDA, which uses statistical models to analyze hundreds of process parameters from sensors in real time. This allows for the detection of subtle drifts from the optimal state, enabling predictive alerts that prevent batch deviations and contamination before they occur.
  • Continuous Manufacturing: Unlike traditional, slow batch-based production, continuous manufacturing runs 24/7. This modern approach is impossible without real-time analytics to monitor Critical Process Parameters (CPPs) and ensure Critical Quality Attributes (CQAs) are consistently met. This leads to a smaller physical footprint, less waste, higher consistency, and enables Real-Time Release Testing (RTRT), where product can be released for distribution based on in-process data, slashing cycle times.
  • Predictive Maintenance & Anomaly Detection: By analyzing data from equipment sensors, machine learning models can predict when a piece of machinery—like a chromatography column or a filling line pump—is likely to fail. This allows maintenance to be scheduled proactively, avoiding costly unplanned downtime and potential batch loss.
  • Supply Chain Forecasting: In an increasingly volatile global environment, a resilient supply chain is a competitive advantage. AI-powered analytics can improve demand forecasting by analyzing market trends, epidemiological data, and customer ordering patterns. It can also optimize global inventory levels and logistics to ensure an uninterrupted supply of medicines to patients.

For a comprehensive look at these applications, read our detailed analysis of advanced analytics in pharma manufacturing and supply chain.

Enhancing Commercial and Post-Market Surveillance

After a drug is launched, analytics remains critical for maximizing its value, ensuring patient safety, and informing the next generation of therapies.

  • Market Access & Pricing: In a value-based healthcare environment, RWE is essential for commercial success. Analytics on large-scale claims and EHR data helps build compelling value propositions for payers, demonstrating a drug’s cost-effectiveness and positive outcomes in the real world.
  • Proactive Pharmacovigilance: Traditional pharmacovigilance is reactive, relying on submitted reports. Modern approaches use AI and NLP to proactively scan millions of unstructured data sources—including adverse event reports (like FAERS), social media, patient forums, and the scientific literature. This allows for the much earlier detection of potential safety signals, protecting patients and enabling a faster response.
  • Social Media Listening: Unsolicited patient feedback on social media and forums provides invaluable real-world insights into how a drug is being used, its perceived benefits, and its side effects. NLP tools can analyze this sentiment to understand the patient experience, identify unmet needs, and detect potential safety issues that may not appear in traditional reporting channels.

Real-time safety surveillance is a competitive advantage and a patient safety imperative. Find how our platform enables Real-time Adverse Drug Reaction Surveillance that protects patients while supporting regulatory compliance.

Building a Scalable and Compliant Analytics Foundation

Unlocking the full potential of biopharma data analytics is impossible without a robust, secure, and compliant foundation. The most sophisticated AI model is useless—and potentially dangerous—if it is built on poor-quality data or operates in a way that violates regulatory requirements. This foundational layer, built on thoughtful governance and a scalable architecture, is what separates successful, sustainable analytics programs from failed science projects.

Secure, federated data architecture diagram - Biopharma data analytics

Data Governance, Regulation, and Ethics

In the highly regulated world of biopharma, data governance is not just an IT function; it is the bedrock of analytics and a prerequisite for trust. It encompasses a framework of principles, regulations, and ethical considerations.

  • FAIR Principles: To maximize the value of data, it must be Findable, Accessible, Interoperable, and Reusable. This means more than just storing data. Findable requires rich metadata and globally unique, persistent identifiers. Accessible means that data (and metadata) are retrievable using standardized protocols, with clear and transparent conditions for access. Interoperable requires the use of common data formats and vocabularies (ontologies) so data can be understood and combined by machines. Reusable means providing clear data usage licenses and detailed provenance to ensure future users know the data’s origin and context.
  • Regulatory Compliance: Biopharma analytics systems operate within a complex web of regulations. GxP requirements (Good Clinical, Laboratory, and Manufacturing Practices) mandate data integrity, which is often defined by the ALCOA+ principles: data must be Attributable, Legible, Contemporaneous, Original, and Accurate, plus Complete, Consistent, Enduring, and Available. Systems must also comply with 21 CFR Part 11 for electronic records and signatures, which requires features like secure audit trails and system validation. On top of this, strict privacy laws like HIPAA in the US and GDPR in Europe govern the use of patient data. Data residency rules, reinforced by rulings like Schrems II, add further complexity, often restricting the transfer of EU patient data to other regions, making federated approaches essential.
  • Ethical AI: As AI makes increasingly critical decisions, ensuring ethical use is paramount. A key challenge is bias mitigation. If an AI model is trained on data from a single demographic, its predictions may be inaccurate or unfair for other populations. This requires careful curation of training data and continuous monitoring for performance disparities. Furthermore, the concept of Explainable AI (XAI) is gaining importance. In a regulated environment, a ‘black box’ model is often unacceptable; researchers, clinicians, and regulators need to understand why a model made a particular prediction to trust and act on it.

Building trust through secure data collaboration is at the heart of everything we do. Learn more about how we approach AI-Enabled Data Governance to balance innovation with compliance.

Designing a Technical Architecture for Biopharma Data Analytics

A future-proof technical architecture must be scalable, flexible, and secure, capable of handling the unique variety, volume, and velocity of biopharma data.

  • Deployment Models: Organizations can choose between cloud, on-premises, and hybrid approaches. While on-premise offers maximum control, the cloud provides unparalleled scalability, access to cutting-edge tools, and cost-efficiency. Hybrid models are increasingly popular, allowing organizations to keep highly sensitive data on-premise while leveraging the massive computational power of the cloud for analysis. A key architectural pattern emerging is federated analytics, where analytical models are securely sent to the data’s location, rather than moving sensitive data. This respects data residency, preserves patient privacy, and enables secure collaboration across institutional and national boundaries.
  • The Data Lakehouse: This modern architecture represents the evolution of data platforms. It combines the low-cost, flexible storage of a data lake (ideal for raw, unstructured data like genomic sequences or medical images) with the performance, management, and transactional features of a data warehouse (ideal for structured clinical or manufacturing data). By unifying these capabilities, a data lakehouse provides a single, consistent platform for all data types, breaking down silos and simplifying the data landscape for AI and analytics workloads.
  • Data Management: Foundational tools for data management are non-negotiable. Metadata management catalogs all data assets, making them findable and understandable. Master Data Management (MDM) creates a single, authoritative ‘golden record’ for critical entities like patients, products, and clinical sites, ensuring consistency across the enterprise.
  • Integration and Operations (MLOps): APIs (Application Programming Interfaces) are the glue that connects disparate systems (like LIMS, MES, and EDC systems). ETL/ELT (Extract, Transform, Load / Extract, Load, Transform) processes are the pipelines that ingest, clean, and prepare raw data for analysis. Critically, MLOps (Machine Learning Operations) brings discipline to the AI lifecycle. It automates and tracks model training, validation, deployment, and monitoring, ensuring that AI models are reproducible, auditable, and compliant—a necessity for using AI in a GxP-regulated context.

For a deeper dive into how these architectural components work together in practice, explore our insights on the Application of Data Lakehouses in Life Sciences.

Integrating Key Data Sources and Standards

The power of analytics comes from integrating diverse data sources to create a comprehensive view. This is a significant technical challenge that requires a commitment to standardization and harmonization.

  • Diverse Data Sources: A modern biopharma analytics platform must ingest and harmonize data from a wide array of sources: Multi-omics (genomics, transcriptomics, proteomics, metabolomics); Operational Systems data from LIMS (Laboratory Information Management Systems), MES (Manufacturing Execution Systems), and EDC (Electronic Data Capture) systems; and Real-World Data, which includes structured data like Electronic Health Records and insurance claims, as well as unstructured data from physician notes, medical imaging, wearables/IoT, and even social media.
  • The Critical Role of Data Standards: Integration is impossible without common standards. CDISC standards (like SDTM and ADaM) are essential for governing the format and structure of clinical trial data, ensuring it is submission-ready. HL7/FHIR (Fast Healthcare Interoperability Resources) is the global standard for exchanging healthcare data, enabling interoperability between clinical and research systems. Medical ontologies and terminologies like SNOMED CT (for clinical terms), LOINC (for lab tests), and ICD (for diagnoses) are crucial for ensuring semantic consistency, so that a concept like ‘Type 2 Diabetes’ is understood uniformly across all datasets.

The quality of your insights is directly proportional to the quality of your data. Robust data curation and harmonization are complex, resource-intensive processes, but they are non-negotiable for generating reliable, trustworthy results. Data harmonization is complex but crucial—learn more about Data Harmonization: Overcoming Challenges to understand how we tackle these technical problems while maintaining data integrity and compliance.

The Future of Intelligent Biopharma

The horizon for biopharma data analytics is one of increasing intelligence, automation, and interconnectedness, where data-driven insights are an inherent part of every process.

Smart factory or digital twin in a biopharma context - Biopharma data analytics

The Rise of AI, Digital Twins, and Automation

The future of biopharma is intelligent and automated. Key trends include:

  • Generative AI: Beyond molecule design, it’s streamlining regulatory submissions and scientific publications.
  • Autonomous Labs: Self-driving labs with robots and AI will run experiments 24/7, accelerating research and reducing human error.
  • Smart Factories: IoT sensors and real-time data will enable continuous production optimization and predictive maintenance.
  • Digital Twins: Virtual replicas of processes, organs, or manufacturing lines will allow for risk-free simulation and optimization.

With this power comes the responsibility of risk and bias management. Ensuring data privacy, addressing algorithmic bias, and maintaining transparency are crucial. The future of AI for Precision Medicine is bright but requires thoughtful ethical governance.

Building the Organization of the Future

Technology alone is not enough. Building the organization of the future requires the right people, processes, and partnerships.

  • Talent: Success requires a diverse team of data stewards, bioinformaticians, data engineers, and MLOps specialists.
  • Culture: Effective change management is critical to foster data literacy and encourage a collaborative, data-driven mindset.
  • Strategy: The build vs. buy vs. partner decision must align with your core strengths and resources. When choosing a partner, prioritize compliance readiness, scalability, integration capabilities, and deep industry expertise.

The future belongs to organizations that combine cutting-edge technology with human expertise and ethical responsibility. Building trust is fundamental. Learn more about best practices for Trusted Data Collaboration.

Frequently Asked Questions about Biopharma Analytics

Let me address the most common questions I hear from organizations considering biopharma data analytics for their operations.

What is biopharma data analytics?

Biopharma data analytics is the application of advanced analytics (statistics, ML, AI) to the vast datasets generated across the drug lifecycle—from R&D and clinical trials to manufacturing and post-market surveillance. Its goal is to accelerate drug development, improve quality, optimize operations, and reduce costs. It is unique due to its reliance on deep domain expertise, the highly regulated environment, and the extreme complexity of biological data.

Where does analytics create the most value in biopharma?

Analytics creates the most significant value in three key areas:

  • R&D Acceleration: By identifying novel targets faster, predicting toxicity early, and designing better molecules, analytics fundamentally changes the economics of drug findy.
  • Manufacturing Efficiency: Real-time monitoring, predictive maintenance, and yield optimization deliver immediate and measurable ROI, often reducing batch failures by 30% or more.
  • Clinical Trial Optimization: Smart patient stratification, site selection, and the use of synthetic control arms dramatically reduce trial timelines and costs.

What are the biggest challenges in implementing biopharma analytics?

The biggest challenges in implementing biopharma analytics are:

  • Data Silos: Data is often trapped in disconnected systems across R&D, manufacturing, and clinical departments, hindering comprehensive analysis.
  • Regulatory Compliance: Navigating the complex landscape of GxP, 21 CFR Part 11, HIPAA, and GDPR requires specialized expertise.
  • Data Quality and Interoperability: Inconsistent, incomplete, and non-standardized data undermines the reliability of any analysis.
  • Talent Gaps: There is a shortage of professionals who combine life sciences knowledge with data science and regulatory expertise.

These challenges are solvable with a strategic approach and the right platform that prioritizes compliance, quality, and interoperability from the start.

Conclusion: From Data to Breakthroughs

The evolution of biopharma data analytics is fundamentally reimagining how medicines are developed. We are moving from reactive guesswork to proactive, predictive decision-making, where vast, interconnected data informs every step. This shift allows us to predict a drug’s success, prevent manufacturing failures, and understand real-world treatment outcomes better than ever before.

A compliant, scalable foundation is non-negotiable. Without robust governance and secure infrastructure, analytics efforts can become liabilities.

This is why we built Lifebit’s federated platform. Our Trusted Research Environment (TRE), Trusted Data Lakehouse (TDL), and R.E.A.L. (Real-time Evidence & Analytics Layer) provide secure access to global biomedical data. With built-in harmonization, advanced AI, and federated governance, we enable real-time insights and secure collaboration while ensuring the highest compliance standards.

The future is connected and intelligent. It’s an era where data flows seamlessly, AI accelerates findy, and patients receive treatments developed faster and more precisely. Embracing this data-driven mindset doesn’t just improve business metrics; it accelerates the creation of life-saving breakthroughs.

Learn how a Trusted Research Environment can accelerate your analytics journey


Federate everything. Move nothing. Discover more.


United Kingdom

4th Floor, 28-29 Threadneedle Street, London EC2R 8AY United Kingdom

USA
228 East 45th Street Suite 9E, New York, NY United States

© 2025 Lifebit Biotech Inc. DBA Lifebit. All rights reserved.

By using this website, you understand the information being presented is provided for informational purposes only and agree to our Cookie Policy and Privacy Policy.