Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT03709680
Other study ID # A5481092
Secondary ID 2021-003444-25
Status Active, not recruiting
Phase Phase 1/Phase 2
First received
Last updated
Start date May 24, 2019
Est. completion date February 25, 2025

Study information

Verified date March 2024
Source Pfizer
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

A study to learn about safety and find out maximum tolerable dose of palbociclib given in combination with chemotherapy (temozolomide with irinotecan or topotecan with cyclophosphamide) in children, adolescents and young adults with recurrent or refractory solid tumors (phase 1). Neuroblastoma tumor specific cohort to further evaluate antitumor activity of palbociclib in combination with topotecan and cyclophosphamide in children, adolescents, and young adults with recurrent or refractory neuroblastoma. Phase 2 to learn about the efficacy of palbociclib in combination with irinotecan and temozolomide when compared with irinotecan and temozolomide alone in the treatment of children, adolescents, and young adults with recurrent or refractory Ewing sarcoma (EWS).


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 128
Est. completion date February 25, 2025
Est. primary completion date June 24, 2024
Accepts healthy volunteers No
Gender All
Age group 2 Years to 20 Years
Eligibility Inclusion: 1. Histologically confirmed relapsed or refractory solid tumor as follows: - For dose escalation and dose determination parts: Histologically confirmed relapsed or refractory solid tumor (including CNS tumors but not lymphomas). Patients with Diffuse Intrinsic Pontine Glioma do not require histological only radiographic confirmed relapse to enroll. - For dose expansion and tumor specific cohorts: Histologically confirmed relapsed or refractory solid tumor including but not limited to EWS, rhabdoid tumor, rhabdomyosarcoma, neuroblastoma, and medulloblastoma. Patients with Diffuse Intrinsic Pontine Glioma do not require histological only radiographic confirmed relapse to enroll. EWS is not eligible for TOPO and CTX tumor-specific cohorts. - For randomized Phase 2 part: Histologically confirmed Ewing sarcoma at diagnosis or at relapse, with presence of EWSR1-ETS or FUS-ETS rearrangement. Histopathology confirmation of both EWSR1-ETS or FUS-ETS rearrangement partners is required OR availability of formalin fixed paraffin embedded (FFPE) tumor tissue sample for central testing. Patient must have relapsed or have refractory disease and at least evaluable disease in at least one site other than bone marrow that can be followed by imaging. 2. Age =2 and <21 years at the time of study entry. 3. Lansky performance status =50% for patients =16 years of age, or Eastern Cooperative Oncology Group (ECOG) 0, 1 or 2 for patients >16 years of age. 4. Adequate bone marrow function. - Absolute neutrophil count =1000/mm3; - Platelet count =75,000/mm3 (transfusion independent, no platelet transfusion in past 7 days prior study entry); - Hemoglobin =8.5 g/dL (transfusion allowed). 5. Adequate renal function: Serum creatinine level based on age/gender must within protocol specified limits. 6. Adequate liver function, including: - Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) =2.5 × upper limit of normal (ULN) or =5 × ULN for age, if attributable to disease involvement of the liver; - Total bilirubin =1.5 × ULN for age, unless the patient has documented Gilbert's syndrome.Patients with documented Gilbert's syndrome are eligible if direct bilirubin is within normal ranges (=ULN). 7. Patients enrolled to Phase 1 portion of the study and tumor specific cohorts must have measurable disease as defined by RECIST version 1.1 or modified RANO criteria for CNS disease or at least evaluable disease by INRC for neuroblastoma. The eligible patients with neuroblastoma must have at least one of the following at the time of study entry: - Measurable tumor by CT or MRI that is avid on MIBG scan or demonstrates increased FDG uptake on PET scan; - Avid lesion on MIBG scan with positive uptake at a minimum of one site; - For disease that is not avid by MIBG-scan, at least one lesion that demonstrates increased FDG uptake on PET scan AND viable neuroblastoma confirmed by current or prior biopsy; - bone marrow involvement with more than 5% neuroblastoma cells in at least one sample from bilateral bone marrow biopsies; - In non MIBG-avid refractory soft tissue disease that does not demonstrate increased FDG uptake; lesion biopsy is required to document the presence of viable neuroblastoma, unless patient has a new soft tissue lesion (radiographic evidence of disease progression). Patients with EWS enrolled to Phase 2 portion of the study are eligible with evaluable disease (eg, bone only disease with no soft tissue component). 8. Recovered to CTCAE Grade =1, or to baseline, from any non-hematological acute toxicities of prior surgery, chemotherapy, immunotherapy, radiotherapy, differentiation therapy or biologic therapy, with the exception of alopecia. 9. Serum/urine pregnancy test (for all girls =8 years of age) negative at screening and at the baseline visit. Exclusion: 1. Phase 1 and tumor specific cohorts: For palbociclib with IRN and TMZ combination, prior treatment with a CDK4/6 inhibitor or progression while on treatment with an IRN-containing regimen that includes TMZ. Patients who have received the combination of IRN and TMZ and did not progress while on these medications are eligible. For patients enrolling in the palbociclib with TOPO and CTX combination, prior treatment with a CDK4/6 inhibitor or progression while on treatment with a TOPO-containing regimen that includes CTX. Patients who have received the combination of TOPO and CTX and did not progress while on these medications are eligible. Phase 2 portion: prior treatment with a CDK4/6 inhibitor or progression while on treatment with an IRN-containing or TMZ-containing regimen. Patients who have received IRN and/or TMZ and did not progress while on these medications are eligible. 2. Prior intolerability to IRN and/or TMZ plus/minus palbociclib with IRN and TMZ combination and prior intolerability to TOPO and/or CTX for TOPO and CTX combination. For patients enrolled in the UK, any contraindication for IRN and/or TMZ treatment, as per the local SmPC. 3. Use of strong cytochrome P450 (CYP) 3A inhibitors or inducers within 12 days of study entry. Patients who are receiving strong uridine diphosphate-glucuronosyl transferase 1A1 (UGT1A1) inhibitors within 12 days of Cycle 1 Day 1 (C1D1) are not eligible for the palbociclib with IRN and TMZ combination. Patients who are receiving strong UGT1A1 inhibitors within 12 days of C1D1 are eligible for the palbociclib with TOPO and CTX combination (See Section 5.7.1 for list of products.) 4. Systemic anti cancer therapy within 2 weeks prior to study entry and 6 weeks for nitrosoureas. 5. Prior irradiation to >50% of the bone marrow (see Appendix 9). 6. Participation in other studies involving investigational drug(s) within 2 weeks or 5 half lives, whichever is longer, prior to study entry. 7. Major surgery within 4 weeks prior to study entry. Surgical biopsies or central line placement are not considered major surgeries. 8. For IRN and TMZ with/without palbociclib combinations: known or suspected hypersensitivity to palbociclib, dacarbazine, IRN and/or TMZ. For combination of palbociclib with TOPO and CTX: known or suspected hypersensitivity to palbociclib, TOPO and/or CTX. 9. Patients with known symptomatic brain tumors or brain metastases and require steroids, unless they have been on a stable or on a decreasing steroid dose for >14 days. 10. Patients with previously diagnosed brain metastases are eligible if they have completed their prior treatment and have recovered from the acute effects of radiation therapy or surgery prior to study entry for these metastases for at least 14 days post radiation and 4 weeks post-surgery and are neurologically stable. 11. Hereditary bone marrow failure disorder. 12. QTc >470 msec. 13. History of clinically significant or uncontrolled cardiac disease, including: - History of or active congestive heart failure; if patient had congestive heart failure resolve and >1 year from resolution, patient will be considered eligible; - Clinically significant ventricular arrhythmia (such as ventricular tachycardia, ventricular fibrillation or Torsades de Pointes); - Diagnosed or suspected congenital or acquired prolonged QT syndrome; - Need for medications known to prolong the QT interval; - Uncorrected hypomagnesemia or hypokalemia because of potential effects on the QT interval; - Left ventricular ejection fraction <50% or shortening fraction <28%. 14. Recent or ongoing clinically significant gastrointestinal disorder that may interfere with absorption of orally administered drugs (eg, gastrectomy). 15. Evidence of serious active or uncontrolled bacterial, fungal or viral infection or known history of hepatitis B virus, hepatitis C virus, or human immunodeficiency virus infection or acquired immunodeficiency syndrome-related illness. Screening for viral hepatitis and HIV is under discretion of investigator unless required by local regulation. 16. Severe acute or chronic medical or laboratory test abnormality that may increase the risk associated with study participation or investigational product administration or may interfere with the interpretation of study results, and in the judgment of the Investigator, would make the patient inappropriate for entry into this study. 17. Investigator site staff members directly involved in the conduct of the study and their family members, site staff members otherwise supervised by the investigator, or patients who are Pfizer employees, including their family members, directly involved in the conduct of the study. 18. Fertile male patients or female patients of childbearing potential who are unwilling or unable to follow contraceptive requirements. 19. Pregnant or breastfeeding women.

Study Design


Intervention

Drug:
Palbociclib
Phase 1: Administered (oral) at 55 mg/m2, 75 mg/m2, or 40 mg/m2, or 95 mg/m2 or 115 mg/m2 on days 1-14 of a 21-day cycle Phase 1 Tumor specific cohort-Neuroblastoma and Phase 2 : Administered (oral) at 75 mg/m2 on days 1-14 of a 21-day cycle
Temozolomide
Phase 1 and Phase 2: Administered at 100 mg/m2 (oral or intravenous), on days 1-5 of a 21-day cycle
Irinotecan
Phase 1 and Phase 2: Administered at 50 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
Topotecan
Phase 1 only : Administered at 0.75 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
Cyclophosphamide
Phase 1 only: Administered at 250 mg/m2 (intravenous), on days 1-5 of a 21-day cycle

Locations

Country Name City State
Argentina Hospital Universitario Austral Pilar Buenos Aires
Belgium Cliniques universitaires Saint-Luc Brussels Bruxelles-capitale, Région DE
Belgium Antwerp University Hospital Edegem Antwerpen
Belgium UZ Gent Gent Oost-vlaanderen
Belgium UZ Leuven Leuven Vlaams-brabant
Brazil Fundação Pio XII - Hospital de Câncer de Barretos Barretos SÃO Paulo
Brazil Hospital Pequeno Príncipe Curitiba Paraná
Brazil Hospital Pequeno Principe / A ssociacao Hospitalar de Protecao a Infancia Curitiba Parana
Brazil Hospital Pequeno Principe / A ssociacao Hospitalar de Protecao a Infancia Curitiba Paraná
Brazil Hospital de Clinicas de Porto Alegre Porto Alegre RIO Grande DO SUL
Brazil Hospital de Clinicas de Porto Alegre Porto Alegre RIO Grande DO SUL
Brazil Instituto Nacional de Câncer - INCA Rio de Janeiro
Brazil Hospital Santa Marcelina São Paulo
Bulgaria UMHAT Tsaritsa Yoanna-ISUL EAD Sofia
Canada Alberta Children's Hospital Calgary Alberta
Canada Stollery Children's Hospital Edmonton Alberta
Canada CHU Sainte-Justine Montreal Quebec
Canada The Hospital for Sick Children Toronto Ontario
Czechia Detska nemocnice FN Brno Brno Brno-mesto
Czechia Fakultni nemocnice v Motole Praha Praha 5
France Institut Bergonié - Centre Régional de Lutte Contre Le Cancer de Bordeaux et Sud Ouest Bordeaux Aquitaine
France Centre Leon Berard Lyon Rhône-alpes
France Centre Leon Berard Lyon Rhône-alpes
France Assistance Publique Hôpitaux de Marseille - Hôpital de la Timone Marseille
France Gustave Roussy Villejuif Val-de-marne
Germany Universitaetsklinikum Erlangen Erlangen Bayern
Germany Universitätsklinikum Essen Essen Nordrhein-westfalen
Hungary Borsod-Abaúj-Zemplén Megyei Központi Kórház és Egyetemi Oktatókórház Miskolc Borsod-abaúj-zemplén
Hungary Pecsi Tudomanyegyetem Klinikai Kozpont Pecs
India Artemis hospital Gurugram Haryana
India All India Institute of Medical Sciences New Delhi Delhi
India Rajiv Gandhi Cancer Institute And Research Centre New Delhi Delhi
Israel Tel-Aviv Sourasky Medical Center Dana-Dwek Children's Hospital Tel Aviv Tell Abib
Italy A.O.Universitaria Meyer Firenze Toscana
Italy Ospedale Pediatrico Bambino Gesù IRCCS Rome Roma
Korea, Republic of National Cancer Center Goyang-si Kyonggi-do
Korea, Republic of Asan Medical Center Seoul Seoul-teukbyeolsi [seoul]
Korea, Republic of Samsung Medical Center Seoul Seoul-teukbyeolsi [seoul]
Korea, Republic of Seoul National University Hospital Seoul Seoul-teukbyeolsi [seoul]
Netherlands Prinses Maxima Centrum voor Kinderoncologie Utrecht
Poland Instytut Matki i Dziecka Warsaw Mazowieckie
Poland Uniwersytecki Szpital Kliniczny im. Jana Mikulicza-Radeckiego we Wroclawiu Wroclaw
Slovakia Detska Fakultna nemocnica s poliklinikou Banska Bystrica Banska Bystrica
Slovakia Fakultna nemocnica s poliklinikou F. D. Roosevelta Banska Bystrica, Oddelenie radiologie Banska Bystrica
Slovakia Institut nuklearnej a molekularnej mediciny Banska Bystrica
Slovakia BIONT, a.s. Bratislava
Slovakia Narodny ustav detskych chorob Bratislava
Slovakia Narodny ustav detskych chorob Bratislava
Spain Hospital Universitari Vall d'Hebron Barcelona Barcelona [barcelona]
Spain Hospital Sant Joan de Déu Esplugues de Llobregat Barcelona [barcelona]
Spain Hospital Infantil Universitario Niño Jesús Madrid Madrid, Comunidad DE
Spain Hospital Universitario Virgen Del Rocio Sevilla
Spain Hospital Universitari i Politecnic La Fe València
Sweden Sahlgrenska Universitetssjukhuset Östra Gothenburg Västra Götalands LÄN [se-14]
Sweden Astrid Lindgrens Barnsjukhus Stockholm
Turkey Hacettepe Universite Hastaneleri Ankara
Turkey Ege Universitesi Hastanesi Izmir I?zmir
United Kingdom Royal Hospital for Children Glasgow Scotland
United Kingdom Leeds General Infirmary Leeds
United Kingdom University College London Hospital London London, CITY OF
United Kingdom University College London Hospital, NHS Foundation Trust London
United Kingdom Royal Manchester Children's Hospital Manchester
United Kingdom The Christie NHS Foundation Trust Manchester Greater Manchester
United Kingdom Royal Victoria Infirmary Newcastle upon Tyne England
United Kingdom Royal Victoria Infirmary Newcastle upon Tyne England
United States University of Michigan Health System Ann Arbor Michigan
United States Children's Healthcare of Atlanta at Egleston Atlanta Georgia
United States Children's Healthcare of Atlanta at Scottish Rite Atlanta Georgia
United States Children's Healthcare of Atlanta, Medical Office Building Atlanta Georgia
United States Children's Hospital Colorado Aurora Colorado
United States Children's Blood and Cancer Center Austin Texas
United States Dell Children's Medical Center Austin Texas
United States Johns Hopkins University Baltimore Maryland
United States Children's of Alabama Birmingham Alabama
United States University of Alabama at Birmingham/Children's of Alabama Birmingham Alabama
United States Dana-Farber Cancer Institute Boston Massachusetts
United States Montefiore Medical Center Bronx New York
United States John R. Oishei Childrens Hospital Buffalo New York
United States Roswell Park Cancer Institute Buffalo New York
United States Carolinas Medical Center/Levine Children's Hospital Charlotte North Carolina
United States Torrence E. Hemby Jr. Pediatric Hematology & Oncology Center Charlotte North Carolina
United States Torrey Hemby Center for Cancer and Blood Disorders Charlotte North Carolina
United States Ann & Robert H. Lurie Children's Hospital of Chicago Chicago Illinois
United States Cincinnati Children's Hospital Medical Center Cincinnati Ohio
United States University Hospitals Cleveland Medical Center Cleveland Ohio
United States Nationwide Children's Hospital Columbus Ohio
United States Nationwide Children's Hospital Columbus Ohio
United States Children's Medical Center Dallas Dallas Texas
United States Cook Children's Medical Center Fort Worth Texas
United States UF Health Shands Hospital Gainesville Florida
United States University of Florida College of Medicine Gainesville Florida
United States Cook Children's H/O Infusion Center Grapevine Texas
United States Hackensack University Medical Center Hackensack New Jersey
United States Connecticut Children's Medical Center Hartford Connecticut
United States Penn State Children's Hospital and Penn State Health Milton S. Hershey Medical Center Hershey Pennsylvania
United States Penn State Health Milton S. Hershey Medical Center Hershey Pennsylvania
United States Kapiolani Medical Center for Women and Children Honolulu Hawaii
United States Texas Children's Hospital Houston Texas
United States Indiana University Indianapolis Indiana
United States University of Mississippi Medical Center Jackson Mississippi
United States Children's Mercy Hospital Kansas City Missouri
United States Cincinnati Children's Liberty Campus Liberty Township Ohio
United States MemorialCare Health System - Long Beach Medical Center Long Beach California
United States Children's Hospital Los Angeles Los Angeles California
United States Norton Children's Hospital Louisville Kentucky
United States Novak Center for Children's Health Louisville Kentucky
United States Children's Wisconsin Milwaukee Wisconsin
United States Medical College of Wisconsin Milwaukee Wisconsin
United States University of Minnesota Minneapolis Minnesota
United States University of Minnesota Masonic Children's Hospital Minneapolis Minnesota
United States University of Minnesota Medical Center, Fairview Minneapolis Minnesota
United States University of Minnesota/Masonic Cancer Center Minneapolis Minnesota
United States Robert Wood Johnson University Hospital New Brunswick New Jersey
United States Rutgers Cancer Institute of New Jersey New Brunswick New Jersey
United States Cohen Children's Medical Center New Hyde Park New York
United States Columbia University Medical Center - Herbert Irving Pavilion New York New York
United States Morgan Stanley Children's Hospital of New York-Presbyetrian Hospital New York New York
United States Children's Hospital of The King's Daughters Norfolk Virginia
United States Children's Hospital and Research Center at Oakland Oakland California
United States University of Oklahoma Health Sciences Center Oklahoma City Oklahoma
United States Children's Hospital of Orange County Orange California
United States Arnold Palmer Hospital for Children Orlando Florida
United States Lucile Packard Children's Hospital Palo Alto California
United States University of Illinois College of Medicine at Peoria Peoria Illinois
United States Buerger Center for Advanced Pediatric Care Philadelphia Pennsylvania
United States Children's Hospital of Philadelphia Philadelphia Pennsylvania
United States Phoenix Children's Hospital Phoenix Arizona
United States UPMC Children's Hospital of Pittsburgh Pittsburgh Pennsylvania
United States Children's Medical Center Plano Plano Texas
United States Oregon Health & Science University Portland Oregon
United States Children's Hospital of Richmond at VCU Richmond Virginia
United States Children's Hospital of Richmond at VCU Richmond Virginia
United States Mayo Clinic in Rochester, Minnesota Rochester Minnesota
United States Washington University School of Medicine Saint Louis Missouri
United States Johns Hopkins All Children's Hospital Saint Petersburg Florida
United States Johns Hopkins All Children's Outpatient Care Center Saint Petersburg Florida
United States Intermountain - Primary Children's Hospital Salt Lake City Utah
United States Primary Children's Hospital Outpatient Services Salt Lake City Utah
United States UCSF Medical Center San Francisco California
United States University of California San Francisco, San Francisco California
United States Seattle Children's Hospital Seattle Washington
United States SUNY Upstate Medical University Syracuse New York
United States Johns Hopkins All Children's Hospital Tampa Florida
United States Children's National Hospital Washington District of Columbia
United States Children's National Medical Center Washington District of Columbia

Sponsors (2)

Lead Sponsor Collaborator
Pfizer Children's Oncology Group (COG)

Countries where clinical trial is conducted

United States,  Argentina,  Belgium,  Brazil,  Bulgaria,  Canada,  Czechia,  France,  Germany,  Hungary,  India,  Israel,  Italy,  Korea, Republic of,  Netherlands,  Poland,  Slovakia,  Spain,  Sweden,  Turkey,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Phase 2 open-label, randomized: Event-free survival (EFS) based on Investigator assessment. EFS is defined as the time from randomization until first event (ie, progression, recurrence following response, second malignancy or death without progression or recurrence). Baseline to Month 24.
Primary Phase 1: First Cycle Dose-Limiting Toxicities (DLT) For Dose Escalation/Determination Part: DLT defined as any of the following events occurring during the first treatment cycle and considered at least possibly-related to study medication:Grade 4 neutropenia lasting greater than 7 days;Grade 4 thrombocytopenia lasting greater than 7 days or need for platelet transfusion for a platelet count of < 20,000 per cubic millimeters twice within a 7-day period;greater than 14-day delay in the start of a subsequent course because of neutropenia or thrombocytopenia;Grade 3 or greater non-hematologic toxicities despite optimal treatment;any Grade 2 or greater non-hematologic toxicity requiring discontinuation or interruption of palbociclib for 7 or more consecutive days during the first cycle or any grade non-hematologic toxicity that delays the start of Cycle 2 by more than 14 days;clinically significant non-hematologic laboratory test abnormality Grade 3 or greater not resolving to Grade 1 or baseline within 7 days. First cycle (cycle length is approximately 21 days)
Primary Phase 1: Dose Expansion Parts: Frequency of adverse events For Dose Expansion and Tumor-Specific Expansion Parts: Adverse events to be reported during treatment and for at least 28 days after last dose. At least 28 days after last dose
Primary Phase 1: Dose Expansion Parts: Percentage of Participants With Complete Response or Partial Response For Dose Expansion and Tumor-Specific Expansion Parts: patients with confirmed Complete Response or Partial Response per Response Evaluation Criteria in Solid Tumors (RECIST, v.1.1) or modified Response Assessment in Neuro-Oncology (RANO) for central nervous system malignancies, or International Neuroblastoma Response Criteria (INRC) for neuroblastoma, during study treatment, assessed approximately every 2 to 4 cycles (each cycle is approximately 21 days). Through the end of treatment (up to at least 28 days after last dose)
Secondary Phase 1 and Phase 2: Frequency of adverse events Adverse events to be reported during treatment and for at least 28 days after last dose. At least 28 days after last dose
Secondary Phase 1 and Phase 2: Percentage of participants with laboratory abnormalities Magnesium, Calcium, Creatinine, Albumin, Alanine aminotransferase, Aspartate aminotransferase, Glucose, Phosphorus, Total Bilirubin, Blood urea nitrogen, Alkaline phosphatase, Sodium, Potassium, Chloride, Platelet Count, White Blood Cell Count (differential), hemoglobin, INR or prothrombin Time, HbA1c At least 28 days after last dose
Secondary Phase 1 and Phase 2: Number of Participants With Clinically Significant Treatment-emergent Electrocardiogram (ECG) Findings Clinically significant ECG findings included: corrected QT (QTc) > 450 ms, QTc >500 ms, change in QTc between 30 and 60 ms, change in QTc greater than or equal to 60 ms. At least 28 days after last dose
Secondary Phase 1 and Phase 2: Number of Participants With Clinically Significant Change From Baseline in Vital Signs systolic and diastolic blood pressure, pulse At least 28 days after last dose
Secondary Phase 2 open-label, randomized: Event-free survival (EFS) assessed by an independent review committee. EFS is defined as the time from randomization until first event (ie, progression, recurrence following response, second malignancy or death without progression or recurrence). Baseline to Month 24.
Secondary Phase 1 and Phase 2: Percentage of Participants With Complete Response or Partial Response Patients with confirmed Complete Response or Partial Response per Response Evaluation Criteria in Solid Tumors (RECIST, v.1.1) or modified Response Assessment in Neuro-Oncology (RANO) for central nervous system malignancies, or International Neuroblastoma Response Criteria (INRC) for neuroblastoma, during study treatment, assessed approximately every 2 to 4 cycles (each cycle is approximately 21 days). Through the end of treatment (up to at least 28 days after last dose)
Secondary Phase 1 and Phase 2: Duration of response (DoR) for Participants Who Achieved Complete Response or Partial Response DoR defined as time from date of first response (Complete Response or Partial Response) in responders to date of progression or death Up to 2 years
Secondary Phase 1 and Phase 2: Progression Free Survival (PFS) PFS defined as time from date of enrollment to earliest date of the death or progressive disease Up to 2 years
Secondary Phase 1 and Phase 2: Overall Survival (OS) OS defined as the time from enrollment to date of death due to any cause. Up to 2 years
Secondary Phase 2 : comparison of PET-CT response assessment to objective response on CT/MRI. PET-CT response assessment will be compared to objective response on MRI/CT, as data permit. up to completion of Cycle 4 ( 12 weeks of therapy)
Secondary Phase 2: the impact of the combination of palbociclib with TMZ and IRN treatment on the quality of life (QoL) of patients with refractory or recurrent EWS. Results of QoL reported by patient at baseline and after 2 and 4 cycles using age-appropriate PROMIS tools will be summarized for both treatment arms, as data permit.
Days of hospitalization will be compared in both treatment arms.
Up to Cycle 5 (completion of 12 weeks of treatment)
Secondary Phase 1 and Phase 2:Palbociclib Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Maximum plasma concentration time (Tmax) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2,5, 6,14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Oral plasma clearance (CL/F) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Maximum plasma concentration time (Tmax) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Oral plasma clearance (CL/F) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Maximum plasma concentration time (Tmax) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Irinotecan (and active metabolites), Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Oral plasma clearance (CL/F) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Oral plasma clearance (CL/F) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Topotecan (and active metabolites), Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Maximum plasma concentration time (Tmax) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Cyclophosphamide Pharmacokinetics, Oral plasma clearance (CL/F) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Cyclophosphamide Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Cyclophosphamide Pharmacokinetics, Maximum plasma concentration time (Tmax) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Cyclophosphamide Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
Secondary Phase 1: Cyclophosphamide Pharmacokinetics, Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) Multiple Dose (assuming steady state is achieved), as data permit PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.
See also
  Status Clinical Trial Phase
Active, not recruiting NCT00750841 - Study of the Effect of Rifampicin on the Pharmacokinetics (PK) of Multiple Doses of Cediranib in Patients With Solid Tumours Phase 1
Withdrawn NCT05419817 - Pembrolizumab With Sitravatinib in Recurrent Endometrial Cancer and Other Solid Tumors With Deficient Mismatch Repair System Phase 2
Completed NCT02828930 - A Study to Determine the Excretion Balance, Pharmacokinetics, Metabolism and Absolute Oral Bioavailability of a Single Oral Dose of [14C]-Labeled Idasanutlin and an Intravenous Tracer Dose of [13C]-Labeled Idasanutlin in a Single Cohort of Participants With Solid Tumors (Malignancies) Phase 1
Completed NCT01197170 - Hormone Receptor Positive Disease Across Solid Tumor Types: A Phase I Study of Single-Agent Hormone Blockade and Combination Approaches With Targeted Agents to Provide Synergy and Overcome Resistance Phase 1
Terminated NCT03225105 - M3541 in Combination With Radiotherapy in Solid Tumors Phase 1
Completed NCT03258515 - A Study to Investigate the Effect of Single Dose of AZD6094 (600 mg) on Cardiac Repolarization in Healthy Volunteers Phase 1
Completed NCT01497925 - Ph 1 Trial of ADI-PEG 20 Plus Docetaxel in Solid Tumors With Emphasis on Prostate Cancer and Non-Small Cell Lung Cancer Phase 1
Completed NCT01878890 - Phase I Dose Escalation Trial of Efavirenz in Solid Tumours or Non-Hodgkin Lymphoma in Therapeutic Failure. Phase 1
Active, not recruiting NCT05059522 - Continued Access Study for Participants Deriving Benefit in Pfizer-Sponsored Avelumab Parent Studies That Are Closing Phase 3
Active, not recruiting NCT03634982 - Dose Escalation of RMC-4630 Monotherapy in Relapsed/Refractory Solid Tumors Phase 1
Recruiting NCT04685226 - A Phase I/II Clinical Trial of ICP-723 in the Treatment of Advanced Solid Tumors Phase 1/Phase 2
Recruiting NCT03175224 - APL-101 Study of Subjects With NSCLC With c-Met EXON 14 Skip Mutations and c-Met Dysregulation Advanced Solid Tumors Phase 2
Recruiting NCT06036121 - A Study of ADRX-0706 in Select Advanced Solid Tumors Phase 1
Active, not recruiting NCT03258151 - Association of Genetic Polymorphisms With Docetaxel-based Chemotherapy Toxicities in Chinese Solid Tumor Patients
Completed NCT01528046 - Metformin in Children With Relapsed or Refractory Solid Tumors Phase 1
Recruiting NCT05325866 - A Study Evaluating Bemarituzumab in Solid Tumors With Fibroblast Growth Factor Receptor 2b (FGFR2b) Overexpression Phase 1/Phase 2
Recruiting NCT04557449 - Study to Test the Safety and Tolerability of PF-07220060 in Participants With Advance Solid Tumors Phase 1/Phase 2
Terminated NCT02890368 - Trial of Intratumoral Injections of TTI-621 in Subjects With Relapsed and Refractory Solid Tumors and Mycosis Fungoides Phase 1
Completed NCT02759640 - A Phase I Trial of HS-10241 in Solid Tumors Phase 1
Completed NCT02279433 - A First-in-human Study to Evaluate the Safety, Tolerability and Pharmacokinetics of DS-6051b Phase 1