17 April 2025 - 4:50pm by CIM | 17 April 2025 - 4:51pm by CIM | ||
---|---|---|---|
Changes to Authorization date | |||
- | + | 2017-11-27 00:00:00 | |
Changes to Reference number | |||
- | In process | + | 176 |
Changes to Clinical sites | |||
- | Not applicable | + | Hermanos Ameijeiras Hospital |
+ | Dra. Iraida Caballero Aguirrechu. Second Degree Specialist in Oncology | ||
+ | Havana | ||
Changes to Recruitment status | |||
- | Pending | + | Recruiting |
Changes to Data sharing plan | |||
- | + | Yes | |
Changes to Date of first enrollment | |||
- | 2017-03-01 05:00:00 | + | 2020-02-19 00:00:00 |
Changes to Description of Data Sharing Plan | |||
- | + | Primary data may be requested from the sponsor once the research is completed. | |
Changes to First name | |||
- | Marta | + | Braulio |
Changes to Midle name | |||
- | + | Francisco | |
Changes to Primary outcome(s) | |||
- | Maximum Tolerated Dose-MTD (Level where included 6 subjects). Measuring time: at the end of stage I. The MTD will use in the stage II (expansion to 3 cohort of patients according to tumor type)
| + | Maximum Tolerable Dose (dose including 6 subjects). Measurement time: at the end of Phase I. This dose will be used in Phase II (expansion to 3 cohorts per tumor type).
|
- | Serious adverse events with causality relationship proved (definitive, very likely, likely) with product administration. Measuring time: every 1 or 4 hours if needed during product administration.
| + | Serious adverse events with a demonstrated causal relationship (definite, very probable, or probable) to product administration. Measurement time: every 4 hours or 1 hour as required during product administration. |
- | + | ||
Changes to Last name | |||
- | Osorio Rodriguez | + | Mestre Fernández |
Changes to Medical Specialty | |||
- | Second Degree Specialist in Oncology | + | First Degree Specialist in Oncology |
Changes to Key secondary outcomes | |||
- | 1. Objective Response Rate (Complete response, Partial response according to RECIST 1.1). Measuring time: at baseline, 4 weeks after last dose of second cycle and, Months 6, 9 and 12.
| + | 1. Objective response rate (CR/PR). Measurement time: At enrollment, 4 weeks after the last dose of the second cycle, and at months 6, 9, and 12.
|
- | 2. Overall Survival (Time from randomization until death from any cause or until last news). Measuring time: 12 months.
| + | 2. Overall survival (OS) (Time from patient enrollment to death, regardless of cause, or to the date of last reported death). Measurement time: month 12.
|
- | 3. Progression-Free Survival-PFS (time from randomization until objective tumor progression or death).Measuring time: 12 months.
| + | 3. Progression-free survival (PFS) (Time from the start of treatment to the date of considered disease progression or death from any cause). Measurement time: month 12.
|
- | 4. Response duration (time from patient’s response until objective tumor progression or death). Measuring time: up to 12 months in patients with objective response.
| + | 4. Duration of response (Time from the date the patient achieves a response with study treatment (CR/PR) to objective disease progression or death). Measurement time: up to month 12 in patients who achieve an objective response (CR/PR) with treatment. Immunological Evaluation
|
- | Immunological response
| + | 1. Expansion of effector T cell populations. Measurement time: baseline, 24 hours after the completion of cycles 1 and 2.
|
- | 1. Expansion of effector T lymphocyte populations. Measurement time: at baseline, 24 hours after cycles 1 and 2.
| + | 2. Natural killer (NK) cells. Measurement time: baseline, 24 hours after the completion of cycles 1 and 2.
|
- | 2. Natural killer cells (NK). Measurement time: at baseline, 24 hours after cycles 1 and 2.
| + | 3. Regulatory T cells (Tregs). Measurement time: baseline, 24 hours after the completion of cycles 1 and 2.
|
- | 3. Regulatory T lymphocytes (Treg). Measurement time: at baseline, 24 hours after cycles 1 and 2.
| + | 4. Memory subpopulations. Measurement time: baseline, 24 hours after the completion of cycles 1 and 2.
|
- | 4. Memory subpopulations. Measurement time: at baseline, 24 hours after cycles 1 and 2.
| + | 5. Activation parameters. Measurement time: baseline, 24 hours after the completion of cycles 1 and 2.
|
- | 5. Activation Parameters. Measurement time: at baseline, 24 hours after cycles 1 and 2.
| + | Pharmacokinetic parameters. The following will be performed in phase II:
|
- | Pharmacokinetic parameters. In stage II:
| + | 1. Bioavailability-F (estimated value using the Monolix system, SAEM algorithm combined with Monte Carlo coding). Measurement time: After the first dose of cycle 1 and at the last dose of cycles 1 and 2, using a sparse data block design with 4 patients per sample collection design, where each patient will have 14 draws.
|
- | 1. Bioavailability-F (estimated value using Monolix system, with SAEM algorithm combined with Monte Carlo code). Measuring time: after first dose of cycle 1 and, after last dose of cycles 1 and 2 with a design of few data in block of 4 patients where every patient has 14 extractions.
| + | 2. Volume of distribution (V) (estimated using the Monolix system, SAEM algorithm combined with Monte Carlo coding). Measurement time: After the first dose of cycle 1 and at the last dose of cycles 1 and 2, using a sparse data block design with 4 patients per sample collection design, where each patient will have 14 draws.
|
- | 2. Volume of distribution -V (estimated value using Monolix system, with SAEM algorithm combined with Monte Carlo code). Measuring time: after first dose of cycle 1 and, after last dose of cycles 1 and 2 with a design of few data in block of 4 patients where every patient has 14 extractions.
| + | 3. Elimination constant (k) (estimated using the Monolix system, SAEM algorithm combined with Monte Carlo coding). Measurement time: After the first dose of cycle 1 and at the last dose of cycles 1 and 2, using a sparse data block design with 4 patients per sample collection design, where each patient will have 14 draws.
|
- | 3. Elimination rate constant -k (estimated value using Monolix system, with SAEM algorithm combined with Monte Carlo code). Measuring time: after first dose of cycle 1 and, after last dose of cycles 1 and 2 with a design of few data in block of 4 patients where every patient has 14 extractions.
| + | 4. Maximum concentration (Cmax) (estimated using the Monolix system, SAEM algorithm combined with Monte Carlo coding). Measurement time: after the first dose of cycle 1 and at the last dose of cycles 1 and 2, using a sparse data design in blocks of 4 patients per sample collection design, where each patient will have 14 samples drawn.
|
- | 4. Maximum Plasma Level -Cmax (estimated value using Monolix system, with SAEM algorithm combined with Monte Carlo code). Measuring time: after first dose of cycle 1 and, after last dose of cycles 1 and 2 with a design of few data in block of 4 patients where every patient has 14 extractions.
| + | 5. Half-life (T 1/2) (estimated using the Monolix system, SAEM algorithm combined with Monte Carlo coding). Measurement time: after the first dose of cycle 1 and at the last dose of cycles 1 and 2, using a sparse data design in blocks of 4 patients per sample collection design, where each patient will have 14 samples drawn.
|
- | 5. Elimination half-life -T 1/2 (estimated value using Monolix system, with SAEM algorithm combined with Monte Carlo code). Measuring time: after first dose of cycle 1 and, after last dose of cycles 1 and 2 with a design of few data in block of 4 patients where every patient has 14 extractions.
| + | 6. Plasma clearance (Cl) (estimated using the Monolix system, SAEM algorithm combined with Monte Carlo coding). Measurement time: After the first dose of cycle 1 and at the last dose of cycles 1 and 2, using a sparse data design in blocks of 4 patients per sample collection design where each patient will have 14 samples drawn. Safety
|
- | 6. Plasma Clearance - Cl (estimated value using Monolix system, with SAEM algorithm combined with Monte Carlo code). Measuring time: after first dose of cycle 1 and, after last dose of cycles 1 and 2 with a design of few data in block of 4 patients where every patient has 14 extractions.
| + | 1. Adverse events-AEs (Occurrence of any AE [Yes, No], description [AE name], duration [time elapsed from start date to end date of AE], intensity [Mild, Moderate, Severe, life-threatening or disabling AE, AE resulting in death], severity [Severe/serious or Not Applicable (NP)], outcome [recovered, improved, persists, sequelae], treatment approach [no change, dose modification, temporary or permanent discontinuation of study treatment], causality [1. Definite, 2. Very likely, 3. Probable, 4. Possible, 5. Unrelated, 6. Unknown], treatment indicated for the AE [medicines indicated to counteract the AE], product batch [batch ID]). Measurement time: Every 1 or 4 hours as needed during product administration, at 4 weeks after the last administration, months 6, 9, and 12.
|
- | Safety
| + | 2. Laboratory-hematological tests (hemoglobin, hematocrit, leukogram with differential, and platelet count). Measurement time: Initial evaluation, at 4 weeks after the last administration, months 6, 9, and 12.
|
- | 1. Adverse Events-AE (Occurrence of any AE [Yes, No], description [name of AE], duration [ time from begin date until end date of AE], intensity [Mild, Moderate, Severe, Life-threatening or disabling AE, Death related to AE], gravity [Serious o Not serious, No proceed (NP)], result [recovered, better, persist, sequelae], attitude [without change, dose modification, temporary interruption, definitive interruption], causality relationship [1.Definitive, 2.Very likely, 3.Likely, 4.Possible, 5.No related, 6.Unknown], treatment indicated for AE [medicines indicated for AE], product batch [batch number]). Measuring time: every 1 or 4 hours if needed during the product administration, 4 weeks after last administration and, months 6, 9 and 12.
| + | 3. Laboratory-hemochemical tests (alkaline phosphatase, SGOT, SGPT, creatinine, total and direct bilirubin). Measurement: Baseline assessment, 4 weeks after the last dose, months 6, 9, and 12.
|
- | 2. Laboratory test-hematology (values of hemoglobin, hematocrit, leukogram with differential and platelets). Measuring time: at baseline, 4 weeks after last dose and, months 6, 9 and 12.
| + | 4. Vital signs (Blood pressure [mmg], Heart rate [min], temperature [0°C]). Measurement time: every 1 or 4 hours as needed during product administration, months 6, 9, and 12.
|
- | 3. Laboratory test- biochemistry (values of alkaline phosphatase, AST [GOT], ALT [GPT], creatinine, complete and direct bilirubin). Measuring time: at baseline, 4 weeks after last dose and, months 6, 9 and 12.
| + | 5. Cardiac function assessment (echocardiogram results). Measurement time: at enrollment, 4 weeks after the end of the last cycle.
|
- | 4. Vitals signs (Blood pressure [mmg], Cardiac frequency [min], temperature [0C]). Measuring time: every 1 or 4 hours if needed during product administration and, months 6, 9 and 12.
| + | 6. Physical examination results (Each system will be evaluated as Normal, Abnormal [depending on the findings], Not Examined [if not performed]). Measurement time: before each dose and 1 hour after, months 6, 9, and 12.
|
- | 5. Cardiac function (Echocardiogram result). Measuring time: at baseline, 4 weeks after finished last cycle.
| + | 7. Antibodies against the mutein (Presence or absence). Measurement time: baseline, 24 hours after the end of cycles 1 and 2.
|
- | 8. Physical exam (every system will be evaluated in normal, abnormal [according to findings], not done [if no examined]). Measuring time: before product administration and y 1 hour later, months6, 9 and 12.
| + | |
- | 6. Antibody against Mutein (Present or not). Measuring time: at baseline, 24 hours after cycles 1 and 2. | + | |
Changes to Intervention(s) | |||
- | Stage I
| + | In Phase I, dose escalation will begin at the lowest dose level and will escalate to the next level when the first patient at the lowest dose level has completed the first 14 doses of treatment or at least 9 doses (70% of the planned dose) in the case of definitive discontinuation for a reason other than the presence of a serious adverse event (SAE), without presenting signs or symptoms that show limiting toxicity and that prevent further treatment. The inclusion of patients in successive dose levels will continue in an ascending manner (1 patient per dose level), under the same criteria. If signs or symptoms that prevent further treatment (TLD: dose-limiting toxicity) appear in a patient at any dose level, 2 more patients will be included at the same dose level. If no TLD events occur, the dose will be escalated to the next level, and if a TLD occurs, the next dose level to be de-escalated will be estimated based on the dose-toxicity model updated with the accumulated information. When six subjects are enrolled at the same dose level, this will be considered the Maximum Tolerable Dose (MTD). If the limiting dose for toxicity is not observed between the prescribed levels, a higher dose, estimated using the dose-toxicity model (2400 U/kg), will be considered. De-escalation may be performed to only one dose level below the specified minimum and escalation to only one dose level above the maximum. If the maximum dose level is reached without DLT events, the enrollment of six patients at the same level will be completed. The occurrence of SAEs of grade ≥4 intensity, with demonstrated causality to the non-alpha IL-2 mutein (definite, very probable, or probable causality), will be considered a DLT. Once Stage I is completed and the MTD is defined, Stage II will be performed, assigning patients to up to three cohorts based on the type of tumor diagnosed (pancreatic/triple-negative, ovarian (platinum-resistant), melanoma/clear cell renal). Patients will be assigned continuously to each cohort.
|
- | Group 1, dose level 1 (Experimental): 3000 U/kg of Il-2 not alpha mutein intravenously in not less than 15 minutes, a dose every 8 hours up to 14 doses (cycle 1). The patient will rest for 9 days and the same number of doses will be administered in the second cycle.
| + | Treatment in Stage I will consist of two cycles of intravenous IL-2 non-alpha mutein. In each cycle, one dose will be administered every 8 hours in no less than 15-30 minutes, up to a maximum of 14 doses (4.6 days or 110 hours). The patient will rest for 9 days and receive the same number of doses in the second cycle.
|
- | Group 2, dose level 2 (Experimental): 6000U/kg of Il-2 not alpha mutein intravenously in not less than 15 minutes, a dose every 8 hours up to 14 doses (cycle 1). The patient will rest for 9 days and the same number of doses will be administered in the second cycle.
| + | In Stage II, patients will initially receive one course of treatment (two cycles) of intravenous IL-2 non-alpha mutein. Each dose will be administered over 30 minutes, one dose every 8 hours, up to a maximum of 14 doses (4.6 days or 110 hours). The interval between cycles will be 9 days.
|
- | Group 3, dose level 3 (Experimental): 12000U/kg of Il-2 not alpha mutein intravenously in not less than 15 minutes, a dose every 8 hours up to 14 doses (cycle 1). The patient will rest for 9 days and the same number of doses will be administered in the second cycle.
| + | Four weeks after completing the first course of treatment, the patient will be evaluated by laboratory and imaging studies. Depending on the response (evidence of at least disease stabilization) and if no adverse events contraindicating the use of Mutein appear, the next course of treatment will be administered 4 weeks after the evaluation (2 months after the last Mutein cycle). Courses may be repeated until a full year of treatment is completed, always following the guidelines described for the second course of treatment.
|
- | Stage II
| + | In Stage I, the primary endpoint will be the occurrence of serious adverse events with a proven causal relationship (definite, very probable, or probable) with the administration of the product, and the dose to be evaluated in the next stage will be determined. In Phase II, the pharmacokinetic profile of the non-alpha IL-2 mutein will be characterized in 25 patients treated with the dose selected in Phase I (1200 U/kg). All patients included, regardless of the study stage, will be evaluated for antitumor effect, overall survival, progression-free survival, duration of response, immune response, immunogenicity, and the safety of the treatment regimen.
|
- | The selected dose will use in 3 cohort of patients according to tumor type. | + | The characterization of the pharmacokinetic profile will allow for estimating bioavailability (F), volume of distribution (V), elimination constant (k), maximum concentration (Cmax), half-life (T 1/2), and plasma clearance (Cl). Sampling will be performed during the first course of treatment, after the first dose of cycle 1, and at the last dose of cycles 1 and 2, using a sparse data design in blocks of 5 patients per sample collection design, so that each patient will receive 9 samples. Objective response will be assessed 4 weeks after the completion of the last dose of the non-alpha IL-2 mutein of the second cycle (course 1). If patients receive more courses of treatment, they will also be assessed at the end of the course. If patients do not receive more than one course of treatment, they will be followed up at months 6 and 9 from inclusion, with study completion at 12 months. Laboratory tests and imaging studies will be performed at each of these evaluations.
|
+ | Administration of the non-alpha IL-2 mutein is expected to be safe, assuming that no more than 40% of patients will experience SAEs of grade ≥4 intensity, with a demonstrated causal relationship (definite, very probable, or probable) with the product administration, and up to 33% will experience SAEs of grade 3 severity. The objective response rate (CR and PR) will be estimated as the primary indicator of effect (during the second phase), and is expected to occur in no less than 15% of patients.
| ||
+ | |||
Changes to Target sample size | |||
- | 66 | + | 64 |
Changes to Middle Name | |||
- | + | Cristina | |
Changes to First Name | |||
- | Pedro | + | Tania |
Changes to Middle Name | |||
- | Camilo | + | |
Changes to Last Name | |||
- | Rodriguez Rodriguez | + | Crombet Ramos |
Changes to Specialty | |||
- | Medical doctor, Second Degree Specialist in Pharmacology | + | Medical doctor, Second Degree Specialist in Immunology |
Changes to Source(s) of monetary or material support | |||
- | Center of Molecular Immunology (CIM), Cuba
| + | Center of Molecular Immunology (CIM), CubaMinistry of Public Health (MINSAP), Cuba |
- | Ministry of Public Health (MINSAP), Cuba | + |
Revision of 17 April 2025 - 4:51pm: