What is a Companion Diagnostic

What Is a Companion Diagnostic? The Tests That Unlock Precision Medicine

Table of Contents

Not every drug works for every patient. In oncology, response rates to targeted therapies often range from 20% to 40% in unselected populations. The majority of patients receive treatments that won’t help them — exposing them to side effects without benefit.

Companion diagnostics change this equation.

These tests identify which patients are likely to respond to a specific drug before treatment begins. They’re the gatekeepers of precision medicine — matching the right drug to the right patient based on measurable biology rather than trial and error.

For biotech companies, companion diagnostics shape development strategy and regulatory pathways. For investors, they represent both competitive moats and development complexity. For patients, they mean receiving treatments more likely to work.

This guide explains what companion diagnostics are, how they work, the regulatory framework governing them, and why they’ve become essential to modern drug development.


Companion Diagnostics: The Basic Definition

A companion diagnostic (CDx) is a medical device — typically an in vitro diagnostic test — that provides information essential for the safe and effective use of a corresponding therapeutic product.

The FDA’s definition: A companion diagnostic is an in vitro diagnostic device that provides information that is essential for the safe and effective use of a corresponding therapeutic product.

In practice, this means a test that must be performed before prescribing a specific drug. The test result determines whether a patient should receive the treatment.


Why Companion Diagnostics Exist

Companion diagnostics solve a fundamental problem in medicine: biological heterogeneity.

The Problem

Patients with the same disease diagnosis often have different underlying biology. A breast cancer patient with HER2 overexpression has fundamentally different tumor biology than one without — even though both have “breast cancer.”

Drugs targeting specific biological mechanisms only work when that mechanism is driving the patient’s disease. Giving a HER2-targeted therapy to a HER2-negative patient provides no benefit and only risk.

The Solution

Companion diagnostics identify patients whose biology matches the drug’s mechanism. This enables:

  • Higher response rates — Treating only patients likely to benefit
  • Better risk-benefit ratios — Avoiding toxicity in non-responders
  • Efficient clinical trials — Enriching studies with likely responders
  • Regulatory approval — Demonstrating efficacy in defined populations
  • Appropriate prescribing — Ensuring drugs reach the right patients

How Companion Diagnostics Work

Companion diagnostics detect specific biomarkers that predict drug response. The process works as follows:

Step 1: Sample Collection

A patient sample is collected — typically tumor tissue from a biopsy, but increasingly blood samples for liquid biopsies.

Step 2: Biomarker Testing

The sample is analyzed for the specific biomarker the companion diagnostic measures. This could be:

  • Gene mutations — DNA sequencing to detect specific alterations
  • Gene amplifications — Measuring extra copies of genes
  • Protein expression — Immunohistochemistry (IHC) to detect protein levels
  • Gene fusions — Identifying chromosomal rearrangements
  • Microsatellite status — Measuring DNA repair deficiency

Step 3: Result Interpretation

The test produces a result — typically positive/negative or a quantitative score — compared against a validated cutoff.

Step 4: Treatment Decision

Based on the result, physicians determine whether to prescribe the corresponding drug:

  • Positive result → Patient eligible for treatment
  • Negative result → Patient should receive alternative therapy

The Regulatory Framework

Companion diagnostics face unique regulatory requirements because they directly influence treatment decisions.

FDA Oversight

In the United States, companion diagnostics are regulated as medical devices by the FDA’s Center for Devices and Radiological Health (CDRH), while the corresponding drugs are regulated by the Center for Drug Evaluation and Research (CDER) or Center for Biologics Evaluation and Research (CBER).

Approval Pathways

Companion diagnostics typically require Premarket Approval (PMA) — the most rigorous FDA pathway for medical devices. This requires:

  • Analytical validation (the test accurately measures what it claims to measure)
  • Clinical validation (the test result predicts clinical outcomes)
  • Clinical utility (using the test improves patient outcomes)

Co-Development Requirement

The FDA expects companion diagnostics to be developed alongside their corresponding drugs. Ideally, both receive approval simultaneously, ensuring the test is available when the drug launches.

FDA guidance states: “If safe and effective use of a therapeutic product depends on an IVD companion diagnostic, the therapeutic product and IVD companion diagnostic should be approved or cleared contemporaneously.”

Labeling Requirements

When a drug is approved with a companion diagnostic:

  • The drug label specifies that testing is required
  • The drug label identifies the approved companion diagnostic by name
  • The companion diagnostic label references the corresponding drug
  • Prescribing the drug without testing is considered off-label use

Types of Companion Diagnostic Designations

The FDA categorizes diagnostic tests related to drug therapy into several categories:

Companion Diagnostic (CDx)

Definition: A test that is essential for the safe and effective use of a corresponding drug.

Requirement: Testing is required — the drug should not be prescribed without it.

Example: HER2 testing before prescribing trastuzumab for breast cancer.

Complementary Diagnostic

Definition: A test that identifies patients who are most likely to benefit, but the drug may still be prescribed without it.

Requirement: Testing is recommended but not required.

Example: PD-L1 testing for some pembrolizumab indications — the drug can be prescribed regardless of PD-L1 status in certain cancers, though testing may inform the decision.

Pharmacogenomic Test

Definition: A test that identifies genetic variations affecting drug metabolism, dosing, or adverse event risk.

Requirement: Varies — may be required, recommended, or informational.

Example: HLA-B*5701 testing before prescribing abacavir (HIV medication) to avoid hypersensitivity reactions.


Landmark Companion Diagnostics

Several companion diagnostics have transformed treatment in their respective diseases:

HER2 Testing + Trastuzumab (Herceptin)

Disease: Breast cancer, gastric cancer

Biomarker: HER2 protein overexpression or gene amplification

Test methods: Immunohistochemistry (IHC), fluorescence in situ hybridization (FISH)

Impact: Transformed HER2-positive breast cancer from aggressive, poor-prognosis disease to highly treatable. Established the companion diagnostic paradigm.

EGFR Mutation Testing + EGFR Inhibitors

Disease: Non-small cell lung cancer (NSCLC)

Biomarker: Activating EGFR mutations (exon 19 deletions, L858R)

Test methods: PCR-based mutation detection, next-generation sequencing

Drugs: Erlotinib, gefitinib, osimertinib, others

Impact: Shifted lung cancer treatment from histology-based to mutation-based approach.

BRAF V600 Testing + BRAF Inhibitors

Disease: Melanoma, colorectal cancer, NSCLC, others

Biomarker: BRAF V600E and V600K mutations

Test methods: PCR, sequencing

Drugs: Vemurafenib, dabrafenib (often combined with MEK inhibitors)

Impact: Enabled targeted therapy for BRAF-mutant cancers across multiple tumor types.

ALK Rearrangement Testing + ALK Inhibitors

Disease: Non-small cell lung cancer

Biomarker: ALK gene fusions

Test methods: FISH, IHC, next-generation sequencing

Drugs: Crizotinib, alectinib, ceritinib, others

Impact: Identified a distinct molecular subtype of lung cancer with high response rates to targeted therapy.

PD-L1 Testing + Checkpoint Inhibitors

Disease: Multiple cancers

Biomarker: PD-L1 protein expression

Test methods: Immunohistochemistry

Drugs: Pembrolizumab, atezolizumab, others

Complexity: Multiple PD-L1 tests exist with different antibodies, scoring systems, and cutoffs — creating challenges for standardization.

MSI/dMMR Testing + Pembrolizumab

Disease: Any solid tumor (tumor-agnostic)

Biomarker: Microsatellite instability-high (MSI-H) or mismatch repair deficiency (dMMR)

Test methods: PCR, IHC, next-generation sequencing

Impact: First tissue-agnostic FDA approval — drug approved based on biomarker regardless of cancer type.


The Companion Diagnostic Development Process

Developing a companion diagnostic alongside a drug is a complex, parallel process:

Early Development (Phase 1-2)

  • Identify candidate biomarkers through preclinical research
  • Develop prototype assays for clinical trial use
  • Validate analytical performance
  • Generate initial clinical correlation data

Pivotal Development (Phase 3)

  • Finalize assay specifications and cutoffs
  • Prospectively test all patients in pivotal trials
  • Demonstrate clinical validity (biomarker predicts outcomes)
  • Establish clinical utility (testing improves outcomes)
  • Prepare PMA submission in parallel with drug NDA/BLA

Regulatory Submission

  • Submit PMA to FDA’s CDRH
  • Coordinate review timeline with drug application
  • Address both analytical and clinical validation
  • Align labeling between drug and diagnostic

Approval and Launch

  • Ideally achieve simultaneous approval
  • Ensure diagnostic availability matches drug launch
  • Establish laboratory testing infrastructure

Challenges in Companion Diagnostic Development

Companion diagnostics add complexity to drug development:

Development Timeline Alignment

Drug and diagnostic development must proceed in parallel. Delays in either program affect both. Diagnostic validation requires clinical trial data that may not be available until late-stage development.

Biomarker Validation

Identifying biomarkers that reliably predict response is difficult. Many candidates fail to validate. The biomarker cutoff (positive vs. negative) significantly impacts clinical utility and must be carefully established.

Testing Standardization

Different test platforms, antibodies, and scoring systems can produce different results. PD-L1 testing illustrates this challenge — multiple tests exist with limited interchangeability.

Tissue Requirements

Many companion diagnostics require tumor tissue, which may be limited or inaccessible. Liquid biopsy approaches are emerging but require separate validation.

Economic Considerations

Developing a companion diagnostic costs tens of millions of dollars. The diagnostic market is much smaller than the drug market, creating commercial challenges for standalone diagnostic companies.

Access and Reimbursement

Even approved companion diagnostics face insurance coverage challenges. Testing delays can delay treatment initiation.


Companion Diagnostics and Clinical Trial Design

Companion diagnostics fundamentally shape how clinical trials are designed:

Biomarker-Selected Trials

Enroll only patients positive for the biomarker. This “enrichment” strategy increases the proportion of responders, improving the probability of demonstrating efficacy.

Advantage: Higher response rates, smaller trials, clearer results

Risk: Drug only approved for biomarker-positive population (potentially limiting market)

All-Comer Trials with Biomarker Stratification

Enroll all patients regardless of biomarker status, but stratify by biomarker for analysis. This allows assessment of both the overall population and biomarker-defined subgroups.

Advantage: Understand drug activity across patient segments

Risk: May fail to show efficacy in the overall population even if a subgroup benefits

Biomarker-Driven Adaptive Designs

Begin enrolling all patients, then adapt enrollment based on emerging data — potentially restricting to biomarker-positive patients if they show greater benefit.

Advantage: Flexibility to optimize strategy as data emerges

Risk: Statistical and operational complexity


The Business of Companion Diagnostics

Companion diagnostics create complex commercial dynamics:

For Drug Companies

Advantages:

  • Higher response rates support approval
  • Clear patient identification aids commercial targeting
  • Premium pricing justified by defined responders
  • Competitive moat if companion diagnostic is proprietary

Challenges:

  • Market limited to biomarker-positive patients
  • Diagnostic development adds cost and complexity
  • Testing delays can slow drug adoption
  • Dependence on diagnostic company execution

For Diagnostic Companies

Advantages:

  • Guaranteed market tied to drug utilization
  • Premium pricing for companion diagnostics
  • Regulatory pathway clarity
  • Partnership opportunities with pharma

Challenges:

  • Development timeline tied to drug program
  • Risk if drug fails
  • Smaller market than drug
  • Potential competition from laboratory-developed tests

Partnership Models

Drug and diagnostic development often involve partnerships:

  • Co-development agreements: Pharma partners with diagnostic company from early development
  • Licensing: Pharma licenses diagnostic technology
  • Acquisition: Pharma acquires diagnostic company or assets
  • Service agreements: Diagnostic company provides testing for clinical trials with option for commercialization

Companion Diagnostics and Investment Analysis

For biotech investors, companion diagnostic strategy significantly impacts risk and opportunity:

Positive Signals

  • Clear biomarker hypothesis with preclinical validation
  • Companion diagnostic development partner identified early
  • Biomarker-selected Phase 2 showing strong enrichment effect
  • Regulatory alignment on companion diagnostic pathway
  • Testing infrastructure feasible for commercial launch

Warning Signs

  • Biomarker strategy unclear or changing
  • No diagnostic development partner
  • Late-stage attempt to identify biomarker after unselected trial disappointment
  • Complex or expensive testing limiting practical adoption
  • Multiple competing biomarkers without clear validation

Due Diligence Questions

  • Is a companion diagnostic required for approval?
  • What is the biomarker prevalence in the target population?
  • How will the diagnostic restrict or define the addressable market?
  • Is diagnostic development on track to support drug launch?
  • What testing infrastructure exists for the biomarker?

The Future of Companion Diagnostics

The companion diagnostic field continues to evolve:

Next-Generation Sequencing Panels

Comprehensive genomic profiling (CGP) panels test for hundreds of genes simultaneously, identifying multiple actionable mutations in a single test. FDA-approved panels like FoundationOne CDx serve as companion diagnostics for multiple drugs.

Liquid Biopsy

Blood-based tests detecting circulating tumor DNA (ctDNA) enable non-invasive biomarker testing. FDA-approved liquid biopsy companion diagnostics now exist for several drugs, with more in development.

Multi-Marker Approaches

Moving beyond single biomarkers to signatures combining multiple markers — gene expression profiles, mutation combinations, or integrated scores — for more precise patient selection.

Tumor-Agnostic Indications

Companion diagnostics supporting biomarker-defined approvals regardless of tumor type — MSI-H, NTRK fusions, tumor mutational burden — are expanding precision medicine across cancers.

Real-Time Monitoring

Companion diagnostics evolving from single pre-treatment tests to longitudinal monitoring — tracking resistance mutations, minimal residual disease, or treatment response over time.


Tracking Companion Diagnostic Developments

Companion diagnostic approvals, new biomarker validations, and testing advances shape competitive dynamics in precision medicine. Staying current requires monitoring:

  • FDA companion diagnostic approvals
  • Drug approvals requiring or recommending testing
  • Clinical trial designs and biomarker strategies
  • Diagnostic technology advances
  • Testing guideline updates

Stay Ahead of Precision Medicine News

At BioMed Nexus, we cover companion diagnostic approvals, biomarker developments, and precision medicine advances across biotech, medtech, and pharma.

Our free daily newsletter delivers essential developments every weekday morning — synthesized into a briefing you can read in minutes.

85,000+ life science professionals trust BioMed Nexus to keep them informed.

Subscribe free to BioMed Nexus


For Complete Strategic Intelligence

If your work requires comprehensive coverage of clinical development strategies, regulatory catalysts, and competitive dynamics — including companion diagnostic approvals reshaping therapeutic areas — BioMed Nexus Premium delivers institutional-grade intelligence daily.

Premium members receive:

  • 30-Day Catalyst Watch — Complete calendar of regulatory events including companion diagnostic approvals
  • Binary Risk Matrices — Structured analysis of upcoming drug approvals, including companion diagnostic requirements
  • Deal Heat Tracking — M&A and partnership activity in precision medicine and diagnostics
  • Premium Strategic Insights — Deep analysis on biomarker strategies, testing requirements, and competitive implications
  • Institutional Positioning — How sophisticated investors evaluate companion diagnostic strategies

Upgrade to BioMed Nexus Premium for complete daily coverage.


The Bottom Line

Companion diagnostics are the gatekeepers of precision medicine. They ensure that targeted therapies reach the patients most likely to benefit — improving outcomes, reducing futile treatment, and enabling drug approvals in defined populations.

For drug developers, companion diagnostics are increasingly mandatory rather than optional. For investors, they represent both risk (development complexity, market restriction) and opportunity (competitive moats, regulatory clarity). For patients, they mean receiving treatments matched to their biology rather than one-size-fits-all approaches.

Understanding companion diagnostics is essential for anyone serious about modern drug development. They’re not peripheral to precision medicine. They’re what makes it possible.

Subscribe to BioMed Nexus — Free


BioMed Nexus delivers daily intelligence for biotech, medtech, and pharma professionals. Trusted by 85,000+ subscribers who stay informed in minutes — not hours.

Featured Articles

Daily Updates

Abbott’s Libre Safety Shock and Itvisma’s Gene Therapy Expansion: A Thanksgiving Reckoning for Device Quality and Rare Disease Access

FDA approves first gene-replacement therapy for SMA patients 2+ years as Abbott’s 3-million sensor correction exposes fundamental questions about CGM accuracy and post-market surveillance The day before Thanksgiving 2025 delivered a stark juxtaposition: medical innovation extending curative treatment to thousands of previously ineligible children with

Read More »

Join 85,000+ Biotech, MedTech, and Pharma Leaders

Your Daily Edge in Biotech, MedTech, and Pharma

Get trusted, high-signal updates every morning
Breakthroughs, trial data, deals, and the news that matters