The largest database of trusted experimental protocols
> Chemicals & Drugs > Organic Chemical > Cabozantinib

Cabozantinib

Cabozantinib: An Optimized Approach to Transformative Cabozantinib Research.
PubCompare.ai harnesses the power of AI to streamline your search for the most effective Cabozantinib protocols from literature, preprints, and patents.
Our innovative comparisons enhance reproducibiltiy and accuracy, ensuring you uncover the optimal Cabozantinib research methods.
Explore PubCompare.ai and elevate your Cabozantinib studies to new heights.

Most cited protocols related to «Cabozantinib»

CheckMate 9ER is a phase 3, randomized, openlabel trial of nivolumab combined with cabozantinib as compared with sunitinib monotherapy. Patients underwent randomization in a 1:1 ratio and were stratified according to IMDC prognostic risk score (0 [favorable] vs. 1 or 2 [intermediate] vs. 3 to 6 [poor]),18 (link),19 (link) geographic region (United States and Europe vs. the rest of the world), and tumor expression of the PD-1 ligand PD-L1 (≥1% vs. <1% or indeterminate). Specific risk factors that make up the IMDC score are included in the Supplementary Appendix, available at NEJM.org. Nivolumab was administered intravenously at a dose of 240 mg every 2 weeks, and cabozantinib was administered orally at a dose of 40 mg once daily. Sunitinib was administered orally at a dose of 50 mg once daily for 4 weeks, followed by 2 weeks off (6-week cycle). All trial treatment continued until disease progression or unacceptable toxic effects, with a maximum 2-year duration of nivolumab treatment. Crossover between groups was not permitted. Dose reductions were not allowed for nivolumab but were permitted for cabozantinib and sunitinib, according to the protocol. Dose delays for adverse events were permitted for all trial drugs. Discontinuation assessments for nivolumab and cabozantinib were made separately for each drug; if discontinuation criteria were met for only one drug, treatment could continue with the other drug that was not related to the observed toxic effect, according to the protocol. Dose-reduction specifications and discontinuation criteria for both groups are detailed in the trial protocol.
Publication 2021
cabozantinib CD274 protein, human Disease Progression Drug Tapering Ligands Neoplasms Nivolumab Patients Pharmaceutical Preparations Sunitinib
As detailed below, a five-step procedure was implemented including (1) elicitation of an initial composite grading algorithm from clinical investigators via a data collection exercise; (2) refinement of the algorithm through targeted questions administered to a broader audience of clinical investigators; (3) use of graphical approaches to ensure the directional consistency of composite grades; (4) quantitative testing of the algorithm’s measurement properties in a national study dataset; and (5) application of the algorithm to data from multi-site randomized cancer clinical trials.
In Step 1 (“Clinical Investigator Grade Assignment”), a data collection form was created with a table showing the 179 possible combinations of PRO-CTCAE item attributes each in a row (e.g., frequency “occasionally”, severity “mild”, interference “somewhat”), with a space for the investigator to select a single composite grade from 0-3 to which he/she felt that combination should map, corresponding to the CTCAE (Table 1, Supplemental Table S2). This form was administered anonymously to 20 clinical investigators with ≥10 years of experience serving as principal or co-investigators for NCI- and industry-sponsored cancer clinical trials. Composite grades assigned by the 20 investigators were tabulated for each combination, with consensus defined a priori as endorsement of a specific numerical grade by ≥75% of investigators for a given combination. Combinations that did not reach consensus were further investigated in Step 2.
In Step 2 (“Clinical Investigator Consensus”), input on the algorithm resulting from Step 1 was systematically elicited from clinical investigators attending the Alliance for Clinical Trials in Oncology biannual group meeting in Chicago, IL. During committee meetings for the Breast, Thoracic, Genitourinary, and Gastrointestinal disease committees, clinical scenarios were presented via PowerPoint slides (Supplemental Figure S1) which described patients with adverse events characterized by various individual attribute combinations. Investigators were asked to vote using electronic audience response units on a single composite grade to which they felt each combination should map, consistent with the CTCAE. No identifying information was collected from the investigators, although there was no overlap between investigators involved in Steps 1 and 2. Clinical scenarios included attribute item score combinations that did not reach consensus in Step 1. Clinical scenarios were also presented for 12 randomly selected item attribute combinations that had reached ≥75% consensus in Step 1 to confirm consensus. Agreement across the respondents was tabulated for each combination. The majority grades were used in the composite grading algorithm evaluated in Step 3.
Step 3 (“Directional Consistency Check”) entailed use of graphical and tabular approaches to ensure the directional consistency of composite grades. Contour plots of composite grades by frequency, severity, and interference were created in Matlab (method developed by co-author Claus Becker) and reviewed to identify whether situations existed such that increasing individual PRO-CTCAE attribute scores would lead to decreasing composite grades. Composite grades for adverse events with two-score combinations (e.g., frequency plus severity) were compared to the range of composite grades for adverse events with three-score combinations (frequency plus severity plus interference) to confirm consistency of composite grades. Directionally inconsistent composite scores were modified and the final composite grading algorithm was then quantitatively tested in Steps 4 and 5.
Step 4 (“Validation”) entailed comparing the measurement properties of the composite grades derived from Steps 1-3 to the previously published measurement properties of the individual items, including validity, reliability, and sensitivity to change. We sought to assess whether measurement properties were comparable. The algorithm was applied to PRO-CTCAE data collected at 2-3 visits in the primary validation study of the PRO-CTCAE,5 (link) which enrolled 940 patients receiving active treatment for a variety of cancer types at 9 US-based centers. Convergent validity was assessed using Pearson correlations between PRO-CTCAE composite grades and European Organisation for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) scales,11 (link) including an overall summary score.12 (link) EORTC QLQ-C30 health-related quality of life summary and functioning/global scales were reverse scored for analysis (original direction retained for graphical representations) such that higher scores represent inferior outcomes, matching the direction of PRO-CTCAE items. Comparison of mean EORTC QLQ-C30 health-related quality of life summary scores across increasing PRO-CTCAE composite grade groups were carried out using Jonckheere-Terpstra tests, which evaluate for monotonically decreasing health-related quality of life for increased PRO-CTCAE composite grade groups.13 Known-groups validity was assessed by comparing mean PRO-CTCAE composite grades between 66 previously defined5 (link) groups of patients on the basis of Eastern Cooperative Oncology Group (ECOG) performance status (0-1 versus 2-4), cancer type, and treatment using two-sample t-tests with effect sizes computed as the differences between group means divided by the pooled standard deviation (i.e., Cohen’s d). Test-retest reliability was estimated using intraclass correlation coefficients based on a one-way analysis of variance model. Sensitivity to change was assessed by comparing changes from first to last visit between groups of patients reporting worsening, no change, or improvement via global impression of change items at the last visit using Jonckheere-Terpstra tests. Standardized response mean was computed per group as the mean change score divided by the standard deviation of the change scores. All methods and measurement properties of individual PRO-CTCAE items have previously been described.5 (link)Finally, in Step 5 (“Clinical Trial Evaluation”), the grading algorithm was applied to PRO-CTCAE data collected in two double-blind placebo-controlled phase III trials: sorafenib vs. placebo in patients with advanced desmoid tumors (Alliance A091105, NCT02066181)14 (link); and cabozantinib vs. mitoxantrone plus prednisone in patients with metastatic castration-resistant prostate cancer (Exelixis COMET-2, NCT01522443).15 Details of patient consent are previously reported.14 (link),15 To assess the utility and meaningfulness of the composite grading approach, individual adverse event rates were compared between arms using individual PRO-CTCAE item scores vs. composite grades. Post-baseline PRO-CTCAE rates were tabulated and compared with Fisher’s exact tests using a previously described method for baseline adjustment,8 (link) following application of the composite grading algorithm to scores at each assessment time point.
Publication 2020
Up to three analyses of the primary end point of overall survival were planned, when approximately 50%, 75%, and 100% of the expected deaths had occurred. We estimated that a sample size of 760 patients, with a total of 621 deaths, would provide the trial with 90% power to detect a hazard ratio of 0.76 favoring cabozantinib over placebo, with a two-sided log-rank test at a 5% level of significance. Assuming a median overall survival of 8.2 months in the placebo group (as shown in the Brivanib Study in HCC Patients at Risk Post Sorafenib [BRISK-PS]28 (link)) and exponential distribution, this would correspond to 32% longer median overall survival (10.8 months) in the cabozantinib group. Inflation of the type 1 error associated with interim analyses was controlled with the use of the Lan–DeMets O’Brien–Fleming alpha spending function.29 (link) If the null hypothesis of no difference in overall survival was rejected at either the first or second interim analysis, testing of secondary end points would proceed, and subsequent analyses of overall survival would not be performed.
Efficacy was assessed in all randomly assigned patients according to the intention-to-treat principle. Safety was assessed in all patients who received at least one dose of the trial regimen. For time-to-event end points, hypothesis testing was performed with the stratified log-rank test with adjustment for the stratification factors used at randomization; median durations and associated 95% confidence intervals were estimated with the Kaplan–Meier method. Hazard ratios were estimated with univariate Cox regression models, with the randomized group as the only predictor. Hazard ratios for overall analyses were calculated from models adjusted for the randomization stratification factors. Hypothesis testing of objective response was performed with the Cochran–Mantel–Haenszel method. All subgroup analyses of overall survival and progression-free survival were prespecified except those based on extrahepatic spread of disease or macrovascular invasion as separate factors and on sorafenib as the only previous therapy. For subgroup analyses, no adjustments were made for multiplicity, and confidence intervals are considered to be descriptive. Hazard ratios for subgroup analyses were calculated from unstratified models except those calculated for the subgroup of patients whose only previous therapy was sorafenib. All analyses were performed with SAS software, version 9.1 or higher (SAS Institute).
Publication 2018
brivanib cabozantinib Patients Placebos Safety Sorafenib Therapeutics Treatment Protocols
Patients were randomly assigned 1:1 to either cabozantinib or everolimus. Randomization was stratified by the number of prior VEGFR-targeting tyrosine kinase inhibitor therapies (1, 2 or more) and risk category per Memorial Sloan-Kettering Cancer Center (MSKCC) prognostic criteria (favorable, intermediate, poor)22 (link) (Fig. 1).
Cabozantinib and everolimus were provided by the sponsor (Exelixis, Inc.), and administered orally daily at 60 mg and 10 mg, respectively. Dose reductions for cabozantinib (40 mg, then 20 mg) and everolimus (5 mg, then 2.5 mg), and interruptions of study treatment were specified for management of adverse events.23 Treatment was continued as long as clinical benefit was observed per investigator, or until development of unacceptable toxicity. Cross over between treatment arms was not allowed.
Publication 2015
Arm, Upper cabozantinib Crossing Over, Genetic Drug Tapering Everolimus FLT1 protein, human Malignant Neoplasms Patient Care Management Patients
The trial was designed to provide adequate power for assessment of both the primary endpoint of progression-free survival and the secondary endpoint of overall survival. For the primary endpoint, 259 events (disease progression or death) are required to provide 90% power to detect a hazard ratio of 0.667 (7.5 months with cabozantinib vs. 5 months with everolimus) using the log-rank test and a two-sided significance level of 5%. For the overall survival endpoint, assuming a single interim analysis at the 33% information fraction at the time of the primary endpoint analysis and a subsequent final analysis, 408 deaths are required to provide 80% power to detect a hazard ratio of 0.75 (20 months with cabozantinib vs. 15 months with everolimus) using the log-rank test and a two-sided significance level of 4%.
Efficacy was evaluated using two populations, each following the intention-to-treat principle. To evaluate the secondary endpoint of overall survival, 650 patients were planned (the overall survival population). However, only 375 patients were required to achieve appropriate statistical power for the primary endpoint of progression-free survival. Thus, the study was designed to evaluate the primary endpoint in the first 375 randomized patients (the progression-free survival population) to allow longer and more robust follow-up of progression-free survival (Fig. 1).
Hypothesis testing for progression-free and overall survival was performed using the stratified log-rank test according to the stratification factors used at randomization. Median duration of progression-free survival and overall survival and associated 95% confidence intervals (CI) for each treatment arm were estimated using the Kaplan-Meier method. Hazard ratios were estimated with a Cox regression model. A prespecified interim analysis for overall survival was conducted at the time of the primary endpoint analysis. The type I error for the interim analysis was controlled by a Lan-DeMets O’Brien-Fleming alpha spending function to account for the actual information fraction at the time of the analysis.
Publication 2015
cabozantinib Disease Progression Everolimus Patients

Most recents protocols related to «Cabozantinib»

Patients were randomized 2:1 to cabozantinib or matching placebo. Randomization was stratified by prior lenvatinib (yes or no) and age (≤65 or >65 years). Patients received a 60 mg tablet of cabozantinib or a matched placebo, which was self-administered orally once per day. All patients received best supportive care, and adverse events (AEs) were managed with dose modification (dose interruptions or reductions) and supportive care. Dose interruptions were allowed for up to 8 weeks or longer with sponsor approval. Dose reductions were from 60 to 40 mg daily and then to 20 mg daily. Patients were treated until disease progression by RECIST version 1.1 or unacceptable toxicity. Treatment beyond disease progression was allowed if patients experienced clinical benefit in the opinion of the investigator. Importantly, patients in the placebo arm who experienced disease progression per blinded independent radiology committee (BIRC) were permitted to crossover and receive open-label cabozantinib.
Publication 2024
CAMILLA is a single-center, open-label, phase I/II multi-cohort trial evaluating cabozantinib plus durvalumab in patients with advanced, treatment-refractory, gastrointestinal malignancies (NCT03539822). The initial completed phase I part of the study has been published21 (link). There are four tumor-specific phase II cohorts of CAMILLA. This is the final report of cohort 2, which consists of patients with unresectable or metastatic CRC. Patients were administered durvalumab 1500 mg intravenously every 28-day cycles and cabozantinib at the recommended phase II dose (RP2D) of 40 mg daily throughout the 28-day cycle. Key study inclusion criteria were the following: (1) advanced or unresectable histologically confirmed colorectal adenocarcinoma; (2) progression or intolerance to at least two prior SOC systemic therapy including progression on an EGFR antibody if tumor is known RAS wild type; and (3) known MSI or MMR status at baseline tumor biopsy. Baseline biopsies were obtained from a metastatic site, which was determined at the discretion of treating investigator. The MSI and MMR testings were only accepted if done in a CLIA-certified lab either locally or through a CLIA-certified commercial vendor. All patients’ tumor samples underwent targeted NGS panel testing as part of SOC practice. This testing covers BRAF and extended RAS mutations which include KRAS and NRAS. Patients were excluded if they had prior treatment with an anti-PD-1, -PD-L1, or -PD-L2 agent or monoclonal antibodies targeting MET receptor or HGF or cabozantinib or other tyrosine kinase inhibitors targeting c-MET. This study was approved by the Kansas University Medical Center (KUMC) Institutional Review Board (IRB) (reference number: IRB1#: IRB00000161). All participants provided written consent at the time of enrollment in accordance with the Declaration of Helsinki.
Publication 2024
Categorical variables were summarized with the number and percentage of the respective group. Quantitative variables were summarized with mean and standard deviation (SD; normally distributed) or median, first, and third quartile (Q1; Q3; non-normally distributed), as specified in the Results Section. Progression-free survival (PFS) times were calculated from the date of initiation of cabozantinib (i.e., the start of the study) until the date of diagnosis of progressive disease (PD), death, or were censored on the date of loss to follow-up or the end of the study (5 February 2022). Overall survival (OS) times were calculated from the date of initiation of cabozantinib (i.e., the start of the study) until the date of death or censored on the date of the end of the study (5 February 2022). Survival times were estimated with the Kaplan–Meier method and compared between the groups using log-rank tests. Cox proportional hazard regression was used to verify the associations between patients’ baseline characteristics, laboratory results, adverse events of ponatinib treatment, and survival (PFS and OS). The multiple Cox models were calculated with a backward stepwise method using the predictors significant in a simple analysis. All the statistical tests were two-tailed, and the results were interpreted as significant at p < 0.05. Statistica software (version 13; Tibco, Tulsa, OK, USA) was used for computations.
Publication 2024
This retrospective analysis included seventy-one patients with biopsy-proven metastatic renal clear cell carcinoma (mRCC) undergoing cabozantinib treatment as a second-line, or further, treatment at the Department of Genitourinary Oncology of the Maria Skłodowska-Curie National Research Institute of Oncology in Warsaw. The database contained the data of patients with mRCC treated at the department between 30 January 2017 and 23 June 2021. This study was performed in line with the principles of the Declaration of Helsinki. Permission to conduct this study was granted by the Maria Sklodowska-Curie National Research Institute of Oncology Bioethics Committee (permission number 38/2018).
Publication 2024
Endpoints presented in this report are PFS (time from randomization to the earlier of either radiographic progression by RECIST version 1.1 per BIRC or death from any cause), objective response rate (ORR; proportion of patients with confirmed complete or partial response by RECIST version 1.1 per BIRC), disease stabilization rate (DSR; proportion of patients achieving a confirmed complete or partial response or stable disease with a duration of at least 16 weeks), and safety and tolerability. While overall survival (OS) was included in COSMIC-311 as an additional endpoint, the study was not powered for OS. Since the primary endpoint was met, and since the study underwent protocol-approved crossover from placebo to cabozantinib, it has been determined that no meaningful results can be obtained by updated OS analyses comparing the cabozantinib arm with the placebo arm.
Efficacy endpoints are presented for prior therapy and histology subgroups and safety endpoints reported for prior therapy subgroups. Prior therapy subgroups were prior sorafenib and not prior lenvatinib (sorafenib only), prior lenvatinib and not prior sorafenib (lenvatinib only), and prior sorafenib and lenvatinib (sorafenib/lenvatinib). Histology subgroups were papillary, follicular, oncocytic (described as Hürthle cell in the primary analysis of the study), and poorly differentiated. Histology determination was performed by investigator. Efficacy and safety endpoints for the overall population have been previously reported.10 (link),11 (link)Tumor response and progression were determined by magnetic resonance imaging or computed tomography at baseline, every 8 weeks for 12 months after randomization, and every 12 weeks thereafter. Safety was assessed every 2 weeks until week 9, every 4 weeks thereafter, and 30 days after treatment discontinuation. AEs were evaluated by investigators with severity graded based on the National Cancer Institute Common Terminology Criteria for Adverse Events, version 5.0.
Publication 2024

Top products related to «Cabozantinib»

Sourced in United States
Cabozantinib is a laboratory reagent used for research purposes. It is a small-molecule tyrosine kinase inhibitor that targets multiple receptor tyrosine kinases involved in tumor growth and angiogenesis. The product is intended for use in scientific research and development activities.
Sourced in United States
Cabozantinib is a laboratory product developed by LC Laboratories. It is a small molecule that inhibits multiple receptor tyrosine kinases.
Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
Sourced in United States, Germany
Crizotinib is a tyrosine kinase inhibitor used in laboratory research. It functions by inhibiting the activity of certain enzymes, such as ALK and c-MET, which are involved in cellular signaling pathways.
Sourced in United States
Cabozantinib is a multi-kinase inhibitor that targets various receptor tyrosine kinases, including MET, VEGFR2, and RET. It is a lab equipment product used in research and scientific applications.
Sourced in United States, Germany, United Kingdom, China, Canada, France, Japan, Australia, Switzerland, Israel, Italy, Belgium, Austria, Spain, Gabon, Ireland, New Zealand, Sweden, Netherlands, Denmark, Brazil, Macao, India, Singapore, Poland, Argentina, Cameroon, Uruguay, Morocco, Panama, Colombia, Holy See (Vatican City State), Hungary, Norway, Portugal, Mexico, Thailand, Palestine, State of, Finland, Moldova, Republic of, Jamaica, Czechia
Penicillin/streptomycin is a commonly used antibiotic solution for cell culture applications. It contains a combination of penicillin and streptomycin, which are broad-spectrum antibiotics that inhibit the growth of both Gram-positive and Gram-negative bacteria.
Sourced in United States, China, Germany, Australia
Sorafenib is a laboratory reagent that functions as a multi-kinase inhibitor. It is commonly used in research settings to study cellular signaling pathways and their modulation.
Sourced in United States, Germany, United Kingdom, Israel, Canada, Austria, Belgium, Poland, Lao People's Democratic Republic, Japan, China, France, Brazil, New Zealand, Switzerland, Sweden, Australia
GraphPad Prism 5 is a data analysis and graphing software. It provides tools for data organization, statistical analysis, and visual representation of results.
Sourced in United States, Germany, United Kingdom, China, Italy, Sao Tome and Principe, France, Macao, India, Canada, Switzerland, Japan, Australia, Spain, Poland, Belgium, Brazil, Czechia, Portugal, Austria, Denmark, Israel, Sweden, Ireland, Hungary, Mexico, Netherlands, Singapore, Indonesia, Slovakia, Cameroon, Norway, Thailand, Chile, Finland, Malaysia, Latvia, New Zealand, Hong Kong, Pakistan, Uruguay, Bangladesh
DMSO is a versatile organic solvent commonly used in laboratory settings. It has a high boiling point, low viscosity, and the ability to dissolve a wide range of polar and non-polar compounds. DMSO's core function is as a solvent, allowing for the effective dissolution and handling of various chemical substances during research and experimentation.
Sourced in United States
Foretinib is a laboratory equipment product designed for use in scientific research and experiments. It serves as a tool for researchers and scientists in their work. The core function of Foretinib is to facilitate specific laboratory procedures and analyses, but a detailed description cannot be provided while maintaining an unbiased and factual approach.

More about "Cabozantinib"

Cabozantinib, also known as XL184 or Cometriq, is a tyrosine kinase inhibitor (TKI) medication used in the treatment of certain types of cancer, including medullary thyroid carcinoma, renal cell carcinoma, and hepatocellular carcinoma.
It works by inhibiting the activity of various tyrosine kinases, such as MET, VEGFR, and RET, which are involved in the growth and spread of cancer cells.
Cabozantinib research is a critical area of study, as it aims to optimize the use of this medication and improve patient outcomes.
PubCompare.ai, an innovative AI-powered platform, can streamline and enhance Cabozantinib research by helping researchers and clinicians easily locate the most effective protocols from the literature, preprints, and patents.
The platform's AI-driven comparisons provide increased reproducibility and accuracy, ensuring that researchers can uncover the optimal Cabozantinib research methods.
This can lead to a better understanding of the drug's mechanism of action, its efficacy in different cancer types, and the identification of potential biomarkers or combination therapies that can further improve its effectiveness.
In addition to Cabozantinib, other related terms and compounds that may be relevant to this research include FBS (fetal bovine serum), which is commonly used in cell culture experiments, Crizotinib, another TKI used in cancer treatment, Penicillin/streptomycin, a commonly used antibiotic combination, Sorafenib, a TKI approved for the treatment of liver and kidney cancer, GraphPad Prism 5, a widely used statistical analysis software, and DMSO (dimethyl sulfoxide), a solvent often used to dissolve compounds in cell-based assays.
By leveraging the power of PubCompare.ai and exploring the diverse range of related terms and compounds, researchers can elevate their Cabozantinib studies to new heights, leading to more effective treatments and improved patient outcomes.