Expertise

Services

Solutions

About Us

Resources

Request Quote

Nonclinical Tox Studies PK TK Analysis Lab Trusted For 20+ Years!

  • Tox Study PK TK Analysis Services At Industry-Leading Turnaround
  • Tailor-Made, High-Value Toxicokinetic Study Bioanalysis Services For Biotech
  • Full-Service Toxicokinetic Analysis From GLP Preclinical Method Development To Clinical Trial Sample Analysis
  • Toxicokinetic (TK) Analysis For Small And Large Molecule Therapeutics On LC-MS/MS And ELISA, Respectively

What Are Toxicology (Tox) Studies?

Nonclinical and In Vivo Toxicology (Tox) Studies: Primary Safety and Toxicity Evaluation for your IND Submission

Nonclinical Toxicology (tox) Studies evaluate drug exposure for adverse effects and therapeutic index during preclinical development. FDA requires toxicological studies in a minimum of two animal species before first in human (FIH) dosing. Generally, one of the selected species for in vivo toxicology studies may be rodent and the other must be non-rodent. Toxicology studies span various designs, including single or acute dose toxicity study, dose range finding (DRF) study, maximum tolerated dose (MTD) study, and repeated dose toxicity studies. We conduct regulated nonclinical tox studies using Good Laboratory Practices (GLP) under 21 CFR part 58. Our toxicology services include routine GLP toxicokinetic (TK) studies and Non-Compartmental Analysis for dose ranging studies. Toxicokinetic (TK) study helps quantitate the systemic exposure time course for prodrugs, drugs, and metabolites. NorthEast BioLab delivers rapid and high-quality non-GLP and GLP toxicity studies to assess your therapeutic compound. Ultimately, acceptable drug safety during tox studies allows FIH testing as your crucial nonclinical toxicology data informs clinical toxicological analysis.

Toxicological or Toxicology (tox) Studies help determine drug safety by evaluating adverse effect profiles, organ and tissue exposures, and margins of efficacy vs. toxicity. Indeed, these toxicology studies in drug development often range doses encompassing the therapeutic index, defining the window between minimum efficacious and maximum tolerated (or feasible) doses. Typically, test articles selected for nonclinical toxicology assessment are optimized drug discovery leads evaluated previously for pharmacokinetics, potency, and efficacy in preclinical disease models. This preliminary data collection is often essential for designing initial toxicology studies, especially for selecting dose levels, routes, and regimens. Tox studies usually begin with dose range-finding (DRF), maximum tolerated dose (MTD), and single ascending dose or short-term (7-day) repeat dose nonclinical toxicology studies. The purpose of these toxicity studies is to ascertain an appropriate dosing regimen for the long term (≥28-day) repeat dose GLP toxicity study. Generally, the repeat dose studies encompass the drug effects on the entire body utilizing various toxicology services such as gross pathology, histology, and microscopy. Furthermore, additional focused studies are performed to evaluate safety on particular organ systems or anticipated risk factors.

Sponsors Love Our Bioanalytical Laboratory Services For Top Quality And Industry Leading Turnaround

Connect With Us For A Reference Call With Them!

NorthEast BioLab is by far the most responsive and thorough bioanalysis lab services CRO.

We partnered on several programs and found NorthEast BioLab responsive, collaborative, and reliable.

We trust NorthEast BioLab to design and execute the most streamlined and impactful bioanalytical projects.

We found their integrity as refreshing as readiness to provide creative scientific input and high-quality data.

We have worked with NorthEast BioLab for over ten years given their commitment to highest quality bioanalytical data.

NorthEast BioLab tremendously supported us in reproducing our critical lab discoveries for drug metabolism.

NorthEast BioLab always exceeds expectations on bioanalytical assay development, validation, and sample analysis.

NorthEast BioLab truly goes that extra mile, and we hope to continue enjoying seminal interactions with them.

NorthEast BioLab presents a science-based, hands-on, no-frills approach on the latest bioanalytical platforms.

We are thrilled to complete our bioanalytical studies with their top quality and incredibly responsive team.

We worked closely to implement the most efficient and cost-effective bioanalytical assay for our PK Studies.

Our projects with NorthEast BioLab include successful method development, validation, stability studies during Clinical Phase I – IV studies.

NorthEast BioLab provides critical insight, and are compliant with regulatory standards and industry best practices. We highly recommend them and look forward to working together again.

Our latest successful study was a pivotal bioequivalence study, where samples from a cross-over study with about 100 volunteers needed swift analysis.

This study, same as all other bioanalytical studies with NorthEast BioLab, was completed with top quality and reporting standard with incredible responsiveness.

As a small company, our vendors are key members of our project teams. The scientists at NorthEast BioLab are technical experts, who produce high-quality data, on-time, and on-budget.

NorthEast BioLab is by far the most responsive and thorough bioanalysis lab services CRO.

We partnered on several programs and found NorthEast BioLab responsive, collaborative, and reliable.

We trust NorthEast BioLab to design and execute the most streamlined and impactful bioanalytical projects.

We found their integrity as refreshing as readiness to provide creative scientific input and high-quality data.

We have worked with NorthEast BioLab for over ten years given their commitment to highest quality bioanalytical data.

NorthEast BioLab tremendously supported us in reproducing our critical lab discoveries for drug metabolism.

NorthEast BioLab always exceeds expectations on bioanalytical assay development, validation, and sample analysis.

NorthEast BioLab truly goes that extra mile, and we hope to continue enjoying seminal interactions with them.

NorthEast BioLab presents a science-based, hands-on, no-frills approach on the latest bioanalytical platforms.

We are thrilled to complete our bioanalytical studies with their top quality and incredibly responsive team.

We worked closely to implement the most efficient and cost-effective bioanalytical assay for our PK Studies.

Our projects with NorthEast BioLab include successful method development, validation, stability studies during Clinical Phase I – IV studies.

NorthEast BioLab provides critical insight, and are compliant with regulatory standards and industry best practices. We highly recommend them and look forward to working together again.

Our latest successful study was a pivotal bioequivalence study, where samples from a cross-over study with about 100 volunteers needed swift analysis.

This study, same as all other bioanalytical studies with NorthEast BioLab, was completed with top quality and reporting standard with incredible responsiveness.

As a small company, our vendors are key members of our project teams. The scientists at NorthEast BioLab are technical experts, who produce high-quality data, on-time, and on-budget.

Tackle Your Nonclinical Toxicology (Tox) Studies With Our Veteran Team Now!

Which Toxicology Services Do You Offer?

Toxicology (Tox) Analysis Services by NorthEast BioLab

NorthEast BioLab provides bioanalysis and pharmacokinetic/toxicokinetic analysis services from early drug discovery through clinical IND/BLA submission and beyond.

Bioanalysis: PK/TK analysis

We offer end-to-end PK/TK analysis services, such as method development, validation, and sample analysis of your formulation and biological samples for test articles, drugs, and metabolites. As needed, our veteran team performs all your preclinical and clinical bioanalytical services using GLP validated equipment, software, and methods. We provide detailed audited study reports for sample bioanalysis and Toxicokinetics as per sponsor requests.

Non-compartmental analysis (NCA): PK/TK parameter analysis

Furthermore, we have in-house expertise to perform your non-compartmental analysis (NCA) – pharmacokinetics or Toxicokinetics of small molecules, metabolites, or monoclonal antibodies during clinical and in vivo preclinical toxicology studies. We can put together Pharmacokinetic Concentration (PC) and Pharmacokinetic Parameter (PP) domains for your results in CDISC SEND or SDTM format for compliance with requirements for electronic submissions to the FDA and other global regulatory agencies.

In line with our solution provider philosophy, we are happy to further assist you with project and regulatory guidance, protocol design, study prosecution, and data interpretation at all project stages. NorthEast BioLab maintains the latest and fully validated software packages for your Toxicokinetic (TK) Study analysis, including Certara’s WinNonlin, Watson LIMS, Sciex Analyst, Spectramax, etc.

Don’t Wait, Start Your Nonclinical Toxicology (Tox) Studies Service Today!

Why Choose Us For Your Tox Studies?

Reliable Toxicokinetic Analysis For Your GLP Tox Studies

At NorthEast BioLab, our veteran team of scientists has the necessary experience and expertise to assist with tox studies from planning and execution to TK analysis and data interpretation. Our expertise during toxicology study would empower you to get toxicokinetic studies right in the first go as an essential component of your drug development process. We perform toxicokinetic analysis to determine systemic exposure, steady-state exposure, and metabolic limitations for your compound. These TK parameters help scientists and clinicians decide the correct drug dosage and dosing regimens for early clinical trials. Hence, it is essential to perform reliable, efficient toxicological studies and analysis to aid compound characterization in drug development. Similarly, we carry out the GLP toxicity studies utilizing a fully validated and reliable bioanalytical and TK method to prepare for regulatory IND submissions. We offer formal and audited bioanalysis and toxicokinetics eCTD reports and present your pharmacokinetic concentration and non-compartmental TK parameter results in a compliant CDISC SEND format.

Complete Guide on IND Enabling Toxicology Studies

In pharmaceutical discovery and development, many drug substances and their formulations are generated. However, the vast majority of these compounds will not be suitable as final products for commercialization. …

What Are The Various Types Of Tox Studies?

Tox Studies: From Nonclinical Toxicology to Clinical Toxicological Analysis

Regulatory authorities mandate toxicological studies for all novel drug entities, their active metabolites, or metabolites demonstrating significant systemic exposure following drug administration. Before performing regulated repeated dose toxicity studies, pharmaceutical and biochemical researchers often perform single dose toxicity study, maximum tolerated dose (MTD) study, and dose range finding (DRF) study to establish appropriate dosing margins. A short term (typically 7 to 14 days) repeated dose toxicity study is often performed to evaluate any acute toxicity and determine whether dose accumulation occurs by performing toxicokinetic (TK) analysis on the first and last days of dosing. These results are then used to design definitive GLP repeat dose toxicology studies, typically dosing for at least 28 days, depending on the drug’s intended clinical administration. Afterward, TK analysis data are generated during these studies to interpret the dose-exposure relationships, whether saturable metabolism occurs with increasing doses or accumulation occurs with multiple doses, and to empirically demonstrate steady-state exposures. Additional evaluations and studies are performed to assess cardiac, pulmonary, or reproductive health effects and the drug’s potential for carcinogenicity or genetic mutations. Assuming the drug of interest demonstrates adequate safety for first in human (FIH) testing, results from the toxicological studies in drug development may be used to aid in the designs for initial single ascending dose (SAD) and subsequent multiple ascending dose (MAD) PK studies in clinical trials.

Trust Our PK TK Analysis Services Lab CRO.

Related FAQs

Answers to additional Toxicology (Tox) Study questions popular among our potential clients.

What are GLP toxicology studies?

In-vivo toxicology studies are required before FIH (First In Human) dosing and are usually performed under GLP conditions using detailed protocols. A general scheme used in the industry for preclinical studies of lead compounds before the commencement of clinical trials is as follows: Acute (single-dose) toxicity dose ranging study, maximum tolerated dose (MTD) study, Repeat-dose toxicity – 2 to 14 weeks (daily dosing, required by regulators in two species, one rodent and one non-rodent), carcinogenicity – up to 2 years of dosing in rodents for chronic treatments, genotoxicity- in vivo mouse micronucleus test, mutagenicity – in vitro AMES test, cardiovascular risk by hERG inhibition, chromosome aberration, and in-vivo respiratory and cardiovascular safety pharmacology studies, as applicable. The majority of these GLP toxicity studies follow regulatory guidance with quality oversight and are performed as toxicology services by CROs.

What is Toxicokinetics (TK) Study?

Toxicokinetic (TK) study is simply a pharmacokinetic study of the drug when administered at quantities higher than the therapeutic dose to evaluate potentially toxic levels of the drug. Thus, Toxicokinetics is the appropriate term for studying the systemic kinetics of all substances at exposure levels ranging from therapy to toxicity. During a GLP toxicity study by FDA standards, the body reaches the maximum tolerated dose (MTD), the dose level yielding the drug’s exposure to the point of toxicity, or the maximum feasible dose (MFD), the highest dose level that can be administered based on physicochemical limitations. Understanding the dosage and systemic exposure of the drug during toxicology studies is essential as this aspect defines the difference in medicine being therapeutic vs. toxic. This difference explains the drug’s therapeutic index, representing the safety margins between therapy (minimum efficacious dose) and toxicity (maximum tolerated dose or maximum feasible dose). The same drug given in a small quantity may start healing at the site of action. But, when administered at a high dose, it can even be fatal for the human body. The necessity for toxicokinetic analysis lies in the study’s ability to provide insights on molecules’ systemic exposure related to adverse effects. This aspect is an incredibly important consideration for the cardiovascular system, the central nervous system, and respiratory assessments. Furthermore, acute single dose toxicity studies carried under this analysis assist in predicting the duration and rate of exposure at the time of one dosing interval and whether systemic accumulation occurs following multiple drug administrations in repeated dose toxicity studies. This benchmark information provides the right dose levels for later stages of the compound development. Overall, the data obtained from toxicokinetic studies assess the toxic responses of various drug compounds. This study’s result becomes the basis for knowing the right starting dose of a new drug that is safe for the human body. Moreover, the analysis may provide insights into how certain medications can diversely affect people of different ages, genders, and ethnicities.

What are the various steps in a Toxicokinetics (TK) Study?

Toxicokinetic analysis is typically performed for acute and repeated dose toxicity studies. It is essentially the process that helps us understand how a substance enters the body and what happens to it inside the body. The term “disposition” is often used in the place of Toxicokinetics to describe the body’s exposure to the drug or toxicant over time. Many factors impact Toxicokinetic studies, but the four critical descriptive underlying processes of disposition are absorption, distribution, metabolism, and elimination. TK analysis mathematically describes these dispositional principles by non-compartmental or compartmental analyses. These stages or processes help learn how the body absorbs, metabolizes, and clears the administered drugs and correlates to clinical toxicological analysis of exposure margins and adverse events for safe clinical administration.

Absorption
Absorption, the first dispositional element in toxicokinetic studies, is the process of a substance entering systemic blood circulation. Depending on the route of administration, drugs may need to overcome some barriers to reach circulation. For example, following oral dosing, a drug must be soluble in the gastrointestinal space and capable of passive or active diffusion across the gut wall. Medicines absorbed through the gastrointestinal tract then reach the portal vein, where first pass metabolism occurs before systemic absorption. In PK and TK studies, the absorption process may be bypassed by administering an intravenous (IV) dose directly into the systemic circulation. Comparison of exposures following extravascular doses (i.e., oral, subcutaneous, dermal, inhaled, etc.) to exposures following IV doses during PK/TK analysis yields bioavailability.

Distribution
Distribution describes the reversible transfer of drugs within the body from one location to another. The distribution of a drug is affected by multiple factors, including lipid-solubility, concentration in plasma and various tissues, and binding to plasma proteins, transport proteins, and tissues. The volume of distribution calculated during PK or TK analysis describes a drug’s ability to enter cells and tissues from systemic circulation.

Metabolism
Metabolism signifies a process by which a drug is converted to other chemical entities (metabolites). Metabolism happens primarily in the liver but can occur throughout the body. At the time of metabolism, the xenobiotic chemicals administered in the body are altered with non-enzymatic or enzymatic reactions. The aim is to convert lipophilic compounds, which are poorly excreted, to more polar entities that can be readily discharged from the body. Understanding drug metabolism rates and sites allow researchers to predict safe exposure levels and potentials for drug-drug or drug-food interactions.

Elimination
Drug chemicals are excreted from the system either as metabolites or in their unchanged form. While the kidney plays an essential role in excreting water-soluble toxins from the body, excretion of other compounds needs the hepatic and biliary systems, etc. During toxicokinetic studies, parameters such as apparent clearance and elimination half-life are calculated to describe compound metabolism and excretion.

What process is involved in Toxicokinetics?

The toxicokinetic process mathematically models the four dispositional factors that act on a compound in a biological system referred to as ADME. They include uptake of the drug into the systemic circulation, or absorption (A), distribution (D) from the bloodstream to cells and tissues within the body, conversion to new entities by biotransformation or metabolism (M), and excretion (E) of the compound and its biotransformation products. Toxicity is often associated with the conversion of the drug compound to more reactive metabolites. We perform toxicokinetic analysis by assessing systemic exposures via bioanalysis of active pharmaceutical ingredients and metabolites, followed by toxicokinetic analysis using the in-vivo time vs. concentration results. Toxicology studies allow determination of i) No observed effect level (NOEL) – the highest dose or exposure that produces no effect, ii) No observed adverse effect level (NOAEL) – the highest dose or exposure that has manageable toxicity, and iii) Therapeutic index (margin of safety) – the range encompassing minimum efficacious dosage to the maximum tolerated dose.

What are the different types of toxicology?

Toxicology CROs offer predictive safety assessments by in-silico and in-vitro methods. In-silico methods allow predictions of potential mutagenicity and genotoxicity based on the compound’s structure and known historical data. In-vitro toxicity assays allow testing with many discovery compounds to predict specific toxicity mechanisms and reduce animal usage. In-vitro toxicity assays test for potential drug-drug interactions (DDI), mutagenicity, cytotoxicity, or organ toxicity using toxicology services including microsome, hepatocyte, hERG, and AMES assays. That said, in-vivo toxicology studies are still critical because they are the ultimate indicator of toxicity. In-vivo toxicity studies include satellite toxicity and maximum tolerated dose (MTD) study assessments, acute single-dose toxicity studies, repeat-dose GLP toxicology studies, genetic and prenatal toxicology, carcinogenic studies, safety pharmacology studies, in-vivo erythrocyte micronucleus tests, and other systems of biology techniques including metabonomics, proteomics, and transcriptomics.

What is nonclinical toxicology?

Nonclinical toxicology and safety pharmacology tests assess a drug’s potential for adverse and undesired effects in animal models before initial testing in humans. The terms nonclinical and preclinical are often used interchangeably since nonclinical tests are evaluated before the inception of clinical trials on novel drug entities.

How is Toxicology used?

We must obtain nonclinical GLP toxicology data for all lead candidates before filing the Investigational new drug application (IND). In-vitro toxicology studies in drug development test mechanisms such as metabolic reactivity, inhibition, induction, mutagenicity, and cytotoxicity. Study results from these assays alert scientists to potential problems or limitations. We perform in-vivo acute and chronic toxicology studies in drug development to optimize and characterize lead discovery candidates. Furthermore, novel techniques including toxicometabonomics, toxicoproteomics, and toxicogenomics can provide higher throughput approaches to identify in-vivo toxic responses soon after dosing or aid in selecting specific endogenous biomarkers for both clinical toxicological analysis and preclinical detection of potential toxicities.

How do you test for drug toxicity?

Before conducting animal testing, toxicity study sponsors must get approval for toxicological studies by the testing organization’s Institutional Animal Care and Use Committee (IACUC). Single-dose or Acute toxicity studies monitor the effect of a single dose on an animal species. In acute toxicological study testing, we administer the investigational product at different doses and observe the results for ~14 days following a single dose. We perform non-GLP repeated dose toxicity studies in rodents (mice or rats) to identify maximum tolerated dose, target tissues, PK, and safety pharmacology profiles during late optimization and before the compound’s preclinical development and nonclinical toxicology assessment. If the test agent has reached a steady-state, and there is a reasonable or no accumulation of test article and no observed acute toxicity at therapeutic levels, then we follow-up with repeat-dose GLP toxicity studies. In repeated dose toxicity studies, the test drug is administered daily for a minimum of 7, 14, 21, or 28 days under GLP conditions. We can also perform subchronic toxicity studies in rodent and non-rodent models with daily dosing for 90 days or more.

What is the difference between TK and PK?

TK analysis is the subdivision of PK to test the relationship between systemic drug exposure and its toxicity in various species. Pharmacokinetic studies determine the systemic exposure of drug administration to living organisms over time after dosing. Toxicokinetic studies are generally carried out with higher doses as compared to pharmacokinetic studies. The objective of toxicokinetic studies is to quantify a compound’s pharmacokinetics in toxicological study species at toxicological doses. TK exposures may increase non-linearly with dosage due to absorption differences, metabolic saturation, or metabolic alterations resulting from the test article’s chronic usage. TK analyses are often used additionally to investigate drug accumulation with repeated dosing, gender differences, and quantitation of known drug metabolites or pharmacodynamic or toxicodynamic biomarkers.

How are pharmacokinetics and Toxicokinetics related?

Pharmacokinetics (PK) encompasses the analysis and description of a drug’s disposition in the body, with a mathematical description of all dispositional processes (ADME – Absorption, Distribution, Metabolism, and Elimination). Toxicokinetic (TK) analysis applies PK principles to the disposition of toxicants and their metabolites related to the time course of toxic or adverse events. PK or TK parameters are determined for the majority of studies using non-compartmental analysis (NCA).

What is SEND and which Tox Studies require data submission in SEND format?

CDER’s SEND (Standard for Exchange of Nonclinical Data) allows for data transfer to the FDA and other national regulatory agencies in a harmonized format allowing for more efficient and standardized regulatory review readable by Janus Nonclinical software. SEND mirrors the Study Data Tabulation Model (SDTM) used for clinical trial results. At present, tox studies fitting in the electronic Common Technical Document format (eCTD) modules 4.2.1.3 (Safety Pharmacology), 4.2.3.1 (Single Dose Toxicity), 4.2.3.2 (Repeat-Dose Toxicity), and 4.2.3.4 (Carcinogenicity) with study start dates on or after March 15, 2019 for NDA/BLA submissions and on or after March 15, 2020 for commercial IND submissions require data submission following the SENDIG 3.1 (SEND Implementation Guide version 3.1). New data standards will be needed for any studies started 24 months after the publication of a Federal Register Notice (FRN) announcing FDA support of the standard and placement on the FDA Data Standards Catalog. The FDA Office of Computational Science (OCS) currently offers a KickStart Program to assist applicants and reviewers in the SEND standard and data transfers to Janus Nonclinical.

Explore More Services