Knowledge

Cold chain test for pharmaceuticals: 2025 compliance guide

How to conduct cold chain tests for pharmaceuticals in 2025

Updated 20 November 2025 — Cold chain test procedures ensure that vaccines, biologics and emerging cell therapies remain safe and potent throughout storage and transport. With the biopharma cold chain market projected to exceed US$65 billion in 2025 and regulations such as the Drug Supply Chain Security Act (DSCSA) requiring interoperable electronic tracking by 2025, effective testing is no longer optional. This comprehensive guide explains why cold chain tests matter, how to perform them, and what trends will shape compliance in 2025 and beyond.

 

Why are cold chain tests essential for pharmaceutical products? – learn about product integrity, market losses and the high failure rate of untested shipments.

Which regulations and standards govern cold chain testing in 2025? – understand DSCSA, FSMA, EU GDP, USP <659> and ICH stability guidelines.

How do you perform temperature mapping and stability testing? – get stepbystep insights into mapping hot and cold spots, using ALCOA+ principles and scheduling tests.

What are DQ, IQ, OQ and PQ validation phases? – discover how packaging, equipment and processes are qualified under realworld conditions.

How do you handle temperature excursions and implement best practices? – explore root causes, corrective actions and emerging technologies for continuous monitoring.

What innovations and trends will shape cold chain testing in 2025? – examine market growth, cell & gene therapy demands, digitalisation and sustainability.

Why cold chain tests matter for pharmaceuticals

Protecting product integrity and patient safety

Cold chain testing safeguards the quality of temperaturesensitive pharmaceuticals by verifying that storage and transport conditions stay within validated ranges. Studies estimate that around 20 % of temperaturesensitive healthcare products are damaged or degraded during distribution due to poor cold chain management. Another report notes that about 30 % of cold chain shipments experience temperature excursions. These failures translate into degraded products, financial losses, regulatory penalties and, most importantly, patient harm.

Key therapeutic categories depend on strict temperature control:

Vaccines and biologics: Most vaccines and monoclonal antibodies must remain between 2 °C and 8 °C, while some require –20 °C or cryogenic storage at –70 °C or lower. The WHO estimates that nearly 50 % of vaccines are wasted globally due to improper temperature management.

Cell and gene therapies: Advanced therapies like CART treatments often demand storage at –150 °C in liquid nitrogen vapour and are forecast to grow from US$6.31 billion in 2024 to US$74.03 billion by 2034. Failure to maintain ultracold conditions can render these treatments ineffective.

Peptide and protein drugs: Diabetes and obesity medications (GLP1 agonists) and coagulation factors require refrigeration and generate demand for specialized logistics.

Without robust testing, even brief exposures outside the approved range can degrade potency and lead to batch rejection. The global biopharmaceutical cold chain market is projected to exceed US$65 billion in 2025, and yet the industry continues to lose billions due to temperature excursions. Cold chain testing provides the evidence needed to demonstrate that systems maintain required conditions, thereby protecting product integrity, ensuring patient safety and reducing waste.

Economic and operational drivers

Beyond patient safety, testing protects revenue streams and reputation. When products are compromised, companies face direct costs (destroyed goods), indirect costs (delayed treatments, rescheduling clinical trials) and reputational damage. Regulatory penalties can include fines, shipment quarantine or licence suspension. With regulatory scrutiny increasing globally, documented evidence of cold chain performance is a competitive advantage for manufacturers, contract development and manufacturing organisations (CDMOs) and logistics providers.

The market for pharmaceutical cold chain services illustrates the growth potential. The global cold chain market for pharmaceuticals was valued at US$6.4 billion in 2024 and is projected to reach US$6.6 billion in 2025. Roots Analysis forecasts growth to US$9.6 billion by 2035, reflecting increasing adoption of biologics and advanced therapies. Meanwhile, healthcare packaging demand is expected to grow over 30 % by 2028. In this context, rigorous cold chain tests serve not only as a compliance requirement but also as a strategic investment.

Regulatory requirements and standards for 2025

Global frameworks and deadlines

Regulators worldwide require proof that temperaturesensitive products are stored and transported within validated ranges. In the United States, the Food Safety Modernization Act (FSMA) emphasises preventive controls and supply chain traceability. The Drug Supply Chain Security Act (DSCSA) mandates interoperable electronic tracking of prescription drugs; dispensers with more than 25 pharmacists must comply by 27 November 2025, while large distributors have deadlines in May and August 2025. The FSMA Food Traceability Final Rule originally scheduled for 2026 may be extended to 20 July 2028, requiring companies handling foods on the FDA’s list to provide key data elements within 24 hours.

Europe’s Good Distribution Practice (GDP) Guidelines and EU GMP Annex 1 demand validated equipment, environmental monitoring and data integrity. The World Health Organization’s Model Guidance on Good Storage and Distribution Practices (WHO TRS 961 Annex 9) and ISO 146443 provide international benchmarks for thermal mapping and cleanroom testing. The International Council for Harmonisation (ICH) Q1 guideline sets out principles for stability testing, stating that the purpose is to provide evidence on how the quality of a drug varies with time under environmental factors such as temperature, humidity, light or agitation and to establish a retest period or shelf life and recommended storage conditions.

In pharmacy practice, the United States Pharmacopeia (USP) General Chapters <659> Packaging and Storage Requirements, <1079> Good Storage and Transportation Practices for Drug Products, and <1079.2> Mean Kinetic Temperature outline requirements for cold chain shipping and call for routine validation and documentation. Accreditation bodies such as URAC and the Accreditation Commission for Health Care (ACHC) include cold chain compliance in their standards; failing to meet them can jeopardise accreditation status.

Data integrity and ALCOA+ principles

Regulators increasingly expect electronic records to comply with ALCOA+ principles—data must be Attributable, Legible, Contemporaneous, Original and Accurate. Electronic record systems must have audit trails, secure user authentication and validated esignatures (21 CFR Part 11). Failing to ensure data integrity can lead to sanctions even when temperature ranges were maintained. When designing cold chain tests, plan for secure data capture, redundant storage and robust access controls.

Temperature mapping and environmental monitoring

Understanding temperature mapping

Temperature mapping, also called thermal mapping or temperature characterisation, measures how temperature is distributed within a controlled space and how it fluctuates over time. Even in a welldesigned cold room or vehicle, temperature is not uniform—it can vary by height, airflow and the presence of equipment. Mapping studies involve placing multiple sensors (typically 9–15 sensors) throughout the area to record temperature data over a defined period. The collected data are analysed to identify the warmest and coldest zones, which then inform the placement of permanent monitoring probes.

The objectives of temperature mapping are to:

Guarantee product quality and storage stability by verifying that the environment stays within validated limits.

Identify hot and cold spots that could compromise product integrity.

Define the best locations for permanent monitoring probes to ensure continuous compliance.

Demonstrate regulatory compliance during audits by providing documented evidence of environmental behaviour.

When and how to conduct mapping

Temperature mapping should occur at several key moments: before initial use of a storage or production area, after significant changes (e.g., HVAC upgrade or equipment replacement) and periodically (often twice a year to account for seasonal variations). Hospitals also map new refrigerators and vaccine storage rooms. Perform mapping both empty (at rest) and loaded to simulate realworld conditions; some teams include stress tests such as door openings and power outages to evaluate system resilience.

Following a mapping study, equip the identified hot and cold points with permanent monitoring probes connected to an integrated Environmental Monitoring System (EMS) that collects, stores, visualises and analyses data in real time. This approach eliminates manual transfers and ensures immediate alerts for deviations.

Data integrity and sensor calibration

To meet ALCOA+ requirements, calibrate sensors against recognised standards (e.g., NIST or UKAS) and document calibration certificates. Use sensors capable of recording temperature, humidity, light and shock events; these should transmit data via secure cellular, satellite or lowpower networks for realtime visibility. Regularly verify sensor placement during mapping and operational phases to avoid blind spots.

Stability testing and shelflife determination

Stability testing evaluates how the quality of a drug substance or product varies with time under the influence of environmental factors and defines its shelf life. ICH Q1 emphasises that stability testing should establish a retest period or expiry period in the proposed container closure system under recommended storage conditions. For cold chain products, stability data not only support label claims but also inform excursion assessments—knowing how long a product can withstand elevated temperatures helps determine whether a shipment remains usable after a deviation.

Stability studies fall into two categories:

Development studies under stress or forced conditions: These characterise degradation pathways and identify critical quality attributes. They may include exposure to elevated temperature and humidity, freeze–thaw cycles and thermal cycling. The results inform analytical methods and help design formal studies.

Formal stability studies (longterm, intermediate and accelerated): These generate data that support shelflife claims and regulatory submissions. For cold chain products, formal studies often include multiple temperature conditions—e.g., 2–8 °C for refrigerated products and –20 °C or –80 °C for frozen and ultrafrozen products. Continuous monitoring and backup power systems are essential to prevent temperature excursions during studies.

Stability tests should be conducted in validated chambers set to ICH stability conditions, and results should be documented according to ALCOA+ principles. Companies often perform additional excursion impact studies to evaluate whether a brief deviation outside the label range affects product quality. These studies allow for sciencebased release decisions when realworld excursions occur.

Validation phases: DQ, IQ, OQ and PQ

Cold chain validation is a structured process comprising four phases—Design Qualification (DQ), Installation Qualification (IQ), Operational Qualification (OQ) and Performance Qualification (PQ). Each phase builds on the previous one and collectively demonstrates that equipment, packaging and procedures perform as intended under realworld conditions.

Design Qualification (DQ) – Evaluate whether the system’s design—vehicles, packaging and monitoring devices—meets the requirements for protecting specific products. Consider materials, insulation thickness, refrigerant type and sensor placement early to avoid costly redesigns.

Installation Qualification (IQ) – Verify that all equipment is installed correctly and functions as intended. For transport, this includes ensuring that refrigeration units, insulated containers and data loggers are set up properly and calibrated. Documentation should include serial numbers, installation diagrams and calibration certificates.

Operational Qualification (OQ) – Test the system under various operating conditions to confirm it performs within specified temperature ranges. Simulate worstcase scenarios such as high ambient temperatures, extended transit times or power failures; verify that alarms trigger at the right thresholds and that the system maintains temperature stability. Temperature mapping is often performed during OQ to identify hot and cold spots.

Performance Qualification (PQ) – Evaluate the system’s performance under actual transport conditions. Conduct pilot shipments along representative routes across seasons and vehicle types. PQ confirms that the system maintains temperatures during storage, loading, transit, unloading and final storage. PQ data provide the confidence needed for commercial deployment.

Validation is not a oneoff exercise. Continuous reassessment is required whenever there are changes in routes, seasons, products or equipment. A validation master plan should outline responsibilities, protocols, acceptance criteria, contingency plans and requalification schedules. Clear documentation of test protocols, results, deviations and corrective actions is critical—without documentation, regulators may deem the test as never having occurred.

Packaging qualification and test planning

In addition to transport systems, the packaging itself must be qualified through the same DQ/IQ/OQ/PQ framework. Qualification testing exposes packages to external forces such as temperature, pressure, humidity and light to ensure they protect contents over the intended transit time. A robust plan should define the test procedure, sample size, test frequency, method justification, aging conditions and acceptance criteria. Only after verification at each level can you progress to the next stage.

Test planning should align with product requirements (e.g., storage time, destination climate, transportation mode) and incorporate ambient temperature profile analysis to select appropriate phasechange materials, insulation and refrigerants. Some packaging suppliers provide offtheshelf operational qualifications, but you still need to perform performance qualifications in your own shipping lanes.

Table: Temperature categories and validation focus

Temperature category Range Typical products Validation focus Practical significance
Refrigerated 2 °C – 8 °C Vaccines, insulin, monoclonal antibodies, GLP1 agonists Verify that passive cooling elements (gel packs, phasechange materials) and insulation maintain the 2–8 °C range throughout the distribution route. Use temperature mapping to locate hot spots. Ensures potency and regulatory compliance; failure leads to product degradation and safety risks.
Frozen Below –20 °C Viral vectors, bulk biologic intermediates, some plasma derivatives Confirm that packaging and refrigeration units can maintain subzero temperatures during loading/unloading; perform drop and vibration tests to ensure physical integrity. Critical for biologics requiring longterm frozen storage; prevents potency loss during transit.
Ultracold/Cryogenic –70 °C to –150 °C mRNA vaccines, cell and gene therapies Validate specialized containers (cryogenic freezers, dry ice shippers) and sensors for extremely low ranges; include realtime tracking and alarm systems. Essential for nextgeneration therapies; failure results in rapid degradation and high financial loss.
Controlled Room Temperature (CRT) 15 °C – 25 °C Many oral drugs, active pharmaceutical ingredients Test that packaging moderates temperature swings and protects against ambient extremes; validate monitoring devices and allow broader excursion ranges supported by stability data. Enables distribution of CRT products without cold chain infrastructure, reducing costs while ensuring safety.
Special categories 8 °C – 15 °C or other narrow bands Certain vaccines, diagnostic reagents Validate packaging for narrow ranges, ensure sensors and alarms provide timely alerts; may involve custom containers and realtime monitoring. Addresses niche products with specific requirements, preventing quality loss in intermediate temperature zones.

Managing temperature excursions and best practices

Understanding excursions and their sources

A temperature excursion occurs when a time–temperaturesensitive product is exposed to temperatures outside its validated storage or transport range. The WHO defines excursions accordingly. Industry data suggest that 20 % of temperaturesensitive products are damaged during distribution, highlighting the prevalence of excursions. Common sources include:

Transportation delays: Traffic congestion, weather disruptions and customs backlogs can prolong transit times.

Packaging failures: Inadequate insulation or depleted phasechange materials lead to rapid temperature changes.

Equipment malfunction: Faulty refrigerators, reefer trucks with inconsistent cooling and inaccurate data loggers cause unexpected deviations.

Human error: Improper loading, doors left open and incorrect configuration of monitoring equipment remain preventable causes.

Regulatory expectations for excursions

Regulators expect a riskbased approach to managing excursions. The U.S. FDA’s GDP guidelines require validated storage and transportation systems capable of maintaining temperature integrity. The European Medicines Agency (EMA) emphasises structured impact assessments and welldocumented responses. The WHO GDP model calls for ongoing monitoring, corrective and preventive actions (CAPA) and robust stability data to support decisions.

Best practices for excursion management

Implementing a comprehensive excursion management strategy involves the following steps:

Develop clear Standard Operating Procedures (SOPs) – SOPs should define every step to take when an excursion occurs, including quarantining affected items, recording temperature and duration, notifying quality assurance teams and conducting root cause analysis. Consistent procedures facilitate audits and regulatory inspections.

Use realtime monitoring and GPS tracking – IoTenabled sensors provide instant alerts for temperature breaches and integrate with GPS to track highrisk shipments. Realtime visibility reduces blind spots and allows rapid corrective actions.

Validate packaging systems and conduct impact studies – Effective packaging reduces the likelihood of excursions. Validate solutions using phasechange materials, vacuuminsulated panels and active or passive containers. Perform stability and excursion impact studies to determine whether excursions affect drug quality and to inform release decisions.

Strengthen workforce training – Human error is a leading cause of excursions, so training should emphasise correct loading/unloading methods, proper use of monitoring equipment and escalation procedures. Regular refresher sessions aligned with GDP changes help maintain competency.

Apply root cause analysis and CAPA – Following an excursion, analyse whether it was caused by equipment failure, packaging, process errors or human error, and implement corrective actions to prevent recurrence.

Leverage emerging technologies – Innovations such as blockchain for immutable records, AIbased predictive analytics to anticipate risks (e.g., weather, customs delays) and smart packaging capable of adjusting thermal profiles are transforming cold chain compliance. Digital dashboards integrate global monitoring, enabling holistic oversight.

Realworld example: COVID19 vaccine distribution

During the COVID19 vaccine rollout, the Pfizer–BioNTech mRNA vaccine required storage at –70 °C. To mitigate excursion risks, the company used GPSenabled thermal shippers, routine dry ice replenishment during transit and continuous digital monitoring from origin to destination. This case underscores how advanced monitoring, validated packaging and proactive risk management ensure safe delivery of ultracold products.

Building a cold chain compliance toolkit

Core risks to address

According to Cold Chain Technologies, a compliant cold chain must mitigate three core risks:

Risk to patient safety: Protect product efficacy, quality and safety.

Risk to total cost of ownership: Avoid the financial and reputational costs of damaged products, recalls and client loss.

Risk to accreditation status: Maintain compliance with URAC, ACHC and other regulatory or accreditation audits.

Risk mitigation checklist

To build your toolkit, start by answering the following questions adapted from industry checklists:

Training: Are your employees and partners trained in URAC and ACHC standards? Do they follow supplier guidelines for packouts? Document training sessions and competencies.

Process review: How often do you review SOPs and packaging procedures? Periodic audits help identify gaps and ensure alignment with current regulations.

Technology use: What temperature monitoring and communication tools are in place? Are you leveraging IoT sensors, blockchain or cloud dashboards to collect and analyse data?

Packaging qualification: Have your packaging systems been qualified to maintain the required temperature range? Do you have performance qualification data for your specific shipping lanes?

Blind spots: Where are the blind spots in your processes, and what are you doing to prevent them?

Patient education: Have patients received correct information about storage, handling and delivery timeframes? Patient communication reduces the risk of exposure to extreme temperatures postdelivery.

Equipment maintenance: When were your freezers and refrigerators last serviced and verified for correct temperature and humidity levels?

Protocol understanding: Do employees understand how refrigeration protocols affect stability and efficacy timelines?

Compliance manual essentials

Create a compliance manual that serves as the blueprint for cold chain success. This manual should include:

Historical testing data: Temperature monitoring and control tests of refrigerators, freezers, performance qualification shipments and realtime product visibility.

Packout diagrams: Seasonal packaging configurations, proper placement of insulation, gel packs, data loggers and other components.

Test summaries: Proof of performance qualification tests conducted in actual shipping lanes, including protocols and results.

Training audits: Summaries of training programs, schedules for refreshers and audits of employee packing and handling procedures.

Performance qualifications: Data and reports submitted to accreditation bodies, along with acceptance criteria and corrective actions.

Standard operating procedures: Cover regulatory compliance, facility and equipment maintenance, inventory management, prescription processing, patient consultation and procedures for detecting and responding to excursions.

Maintaining both physical and digital versions of the manual ensures accessibility and preserves institutional knowledge.

2025 innovations and trends in cold chain testing

Market growth and diversification

The global healthcare cold chain logistics market is projected to grow from US$59.97 billion in 2024 to US$65.14 billion in 2025, with forecasts reaching US$137.13 billion by 2034. The pharmaceutical cold chain services market stands at US$6.6 billion in 2025, and healthcare packaging demand is expected to increase by over 30 % by 2028.

Drivers include the rapid expansion of biologics and biosimilars—more than 85 % of biologics require cold chain management—and the rise of cell and gene therapies requiring cryogenic storage. The number of peptidebased treatments and specialty pharmaceuticals continues to grow, further increasing cold chain complexity.

Digitisation and realtime visibility

In 2025, digital technologies enable unprecedented visibility across the cold chain. IoT sensors integrated into packaging and vehicles continuously measure temperature, humidity, light and shock events. Unlike traditional data loggers that only record data for later analysis, these sensors transmit information instantly via cellular, satellite or lowpower networks, allowing immediate corrective action when deviations occur.

Blockchain solutions offer tamperproof records of temperature measurements and custody transfers, enhancing traceability for highvalue products. AIpowered predictive analytics evaluate factors such as weather patterns, customs delays and carrier reliability to anticipate risks and suggest alternative routes. Digital twins simulate shipments under different conditions, helping to optimise packaging and routing before goods are dispatched.

Sustainable and reusable packaging

Environmental considerations are shaping cold chain innovation. Manufacturers are adopting biodegradable and recyclable materials to reduce waste, and reusable insulated containers are gaining popularity. Vacuuminsulated panels (VIPs) and advanced phasechange materials offer high thermal performance with lower weight, reducing fuel consumption and emissions. Sustainability commitments also drive investments in renewable energy and lowcarbon refrigeration systems.

Regulatory evolution and traceability

Regulatory frameworks will continue to evolve. The DSCSA’s packagelevel electronic tracking requirements become fully enforceable in 2025, and the FSMA Food Traceability Final Rule may shift compliance deadlines to 2028. The EU’s revised GDP guidelines emphasise environmental sustainability, and WHO guidance increasingly integrates riskbased approaches and digital monitoring. Companies must stay current with global standards and harmonise practices across regions.

Integration of quality and supply chain functions

Cold chain testing is no longer solely the domain of quality assurance. In 2025, crossfunctional integration is essential: quality, operations, logistics, IT, sustainability and regulatory teams must collaborate to design, implement and monitor endtoend cold chain solutions. CDMOs with specialised cold chain capabilities are becoming indispensable partners; their integrated infrastructure, specialised equipment, realtime monitoring systems and validated processes support complex biologics and advanced therapies. Supply chain integration ensures seamless coordination from manufacturing to distribution, including clinical trial sites and commercial channels.

Frequently asked questions

Q1: What temperature range do most vaccines require?
Most vaccines and biologics must remain between 2 °C and 8 °C. Keeping products within this controlled cold range preserves potency and meets Good Distribution Practice requirements. Deviations can degrade the active ingredient and lead to batch rejection.

Q2: How often should temperature mapping be performed?
Temperature mapping should occur before initial use, after any major change (e.g., HVAC upgrades) and periodically, typically twice a year to account for seasonal variations. Additional mappings are recommended after changes in product load or room layout.

Q3: What are the main phases of cold chain validation?
Cold chain validation consists of Design Qualification (DQ), Installation Qualification (IQ), Operational Qualification (OQ) and Performance Qualification (PQ). These phases evaluate design suitability, proper installation, operational performance under stress and realworld performance across transport lanes.

Q4: Why is realtime monitoring important?
Realtime monitoring provides instant alerts for temperature breaches and integrates GPS tracking for highrisk shipments. Unlike traditional data loggers, IoT sensors transmit data continuously, enabling rapid corrective action and enhancing regulatory compliance.

Q5: How do you handle a temperature excursion during transport?
Immediately quarantine the product, record temperature and duration, notify quality assurance and perform root cause analysis. Use stability and excursion impact studies to assess whether the product remains within acceptable quality parameters. Document the event and corrective actions thoroughly for regulatory review.

Q6: What innovations are emerging for cold chain testing?
Emerging innovations include blockchain for tamperproof records, AIpowered predictive analytics to anticipate risks, smart packaging that adapts to environmental changes and digital dashboards that integrate data from sensors across global supply chains. These technologies enhance visibility, traceability and risk management.

Conclusion and recommendations

Key takeaways

Testing protects product integrity and patient safety. Around 20 % of temperaturesensitive products are damaged during distribution, and half of vaccines are wasted due to improper temperature management.

Regulatory compliance is nonnegotiable. 2025 brings DSCSA deadlines, evolving GDP guidelines and stricter electronic record requirements. Understanding these frameworks is essential.

Temperature mapping and stability studies are fundamental. Mapping identifies hot and cold spots and informs monitoring probe placement, while stability testing establishes shelf life and supports excursion assessments.

Validation requires a phased approach. Design, installation, operational and performance qualification ensure systems meet design specifications and perform under realworld conditions.

Excursion management and digital tools reduce risk. SOPs, realtime monitoring, validated packaging, training and CAPA processes prevent and mitigate excursions, while IoT, blockchain and AI improve visibility and decisionmaking.

Actionable next steps

Develop or update your validation master plan. Include DQ/IQ/OQ/PQ protocols, risk assessments, acceptance criteria and requalification schedules. Ensure that all sensors and equipment are calibrated and that data integrity controls meet ALCOA+ standards.

Perform comprehensive temperature mapping. Map storage areas and transport vehicles both empty and loaded, twice per year and after major changes. Use the findings to reposition sensors and adjust packaging strategies.

Strengthen excursion management. Create SOPs for excursions, invest in realtime monitoring and conduct stability and excursion impact studies. Train staff on proper loading, packaging and response protocols.

Build a compliance toolkit. Develop a compliance manual with historical test data, packout diagrams, test summaries, training audits and SOPs. Use a digital dashboard to centralise documentation and monitor regulatory changes.

Stay ahead of trends. Monitor emerging regulations, invest in sustainable packaging and adopt digital innovations such as blockchain and AI. Collaborate across quality, logistics and IT functions to integrate cold chain testing into your broader supply chain strategy.

About Tempk

Tempk is a leading provider of thermal packaging and monitoring solutions for the life sciences industry. We design and manufacture insulated shippers, phasechange materials, cryogenic freezers and IoTenabled data loggers that help pharmaceutical companies, CDMOs and logistics providers maintain strict temperature control. Our solutions are validated to meet global GDP and GMP requirements, and our digital platforms integrate realtime monitoring, blockchain traceability and predictive analytics. By partnering with Tempk, you gain access to cuttingedge technology, expert guidance and a commitment to sustainability that ensures your cold chain stays compliant and reliable.

Get in touch to discuss how our cold chain testing and packaging solutions can help you achieve 2025 compliance and beyond.

Previous: Cold Chain Temperature Monitoring Solution Guide 2025 Next: Cold Chain Transportation Services: Keep Perishables Fresh in 2025