The classic website will no longer be available as of June 25, 2024. Please use the modernized ClinicalTrials.gov.
Working…
ClinicalTrials.gov
ClinicalTrials.gov Menu

Study of Onivyde With Talazoparib or Temozolomide in Children With Recurrent Solid Tumors and Ewing Sarcoma

The safety and scientific validity of this study is the responsibility of the study sponsor and investigators. Listing a study does not mean it has been evaluated by the U.S. Federal Government. Know the risks and potential benefits of clinical studies and talk to your health care provider before participating. Read our disclaimer for details.
 
ClinicalTrials.gov Identifier: NCT04901702
Recruitment Status : Recruiting
First Posted : May 25, 2021
Last Update Posted : December 12, 2023
Sponsor:
Collaborators:
Pfizer
Ipsen
Information provided by (Responsible Party):
St. Jude Children's Research Hospital

Brief Summary:

The phase I portion of this study is designed for children or adolescents and young adults (AYA) with a diagnosis of a solid tumor that has recurred (come back after treatment) or is refractory (never completely went away). The trial will test 2 combinations of therapy and participants will be randomly assigned to either Arm A or Arm B. The purpose of the phase I study is to determine the highest tolerable doses of the combinations of treatment given in each Arm.

In Arm A, children and AYAs with recurrent or refractory solid tumors will receive 2 medications called Onivyde and talazoparib. Onivyde works by damaging the DNA of the cancer cell and talazoparib works by blocking the repair of the DNA once the cancer cell is damaged. By damaging the tumor DNA and blocking the repair, the cancer cells may die. In Arm B, children and AYAs with recurrent or refractory solid tumors will receive 2 medications called Onivyde and temozolomide. Both of these medications work by damaging the DNA of the cancer call which may cause the tumor(s) to die.

Once the highest doses are reached in Arm A and Arm B, then "expansion Arms" will open. An expansion arm treats more children and AYAs with recurrent or refractory solid tumors at the highest doses achieved in the phase I study. The goal of the expansion arms is to see if the tumors go away in children and AYAs with recurrent or refractory solid tumors. There will be 3 "expansion Arms". In Arm A1, children and AYAs with recurrent or refractory solid tumors (excluding Ewing sarcoma) will receive Onivyde and talazoparib. In Arm A2, children and AYAs with recurrent or refractory solid tumors, whose tumors have a problem with repairing DNA (identified by their doctor), will receive Onivyde and talazoparib. In Arm B1, children and AYAs with recurrent or refractory solid tumors (excluding Ewing sarcoma) will receive Onivyde and temozolomide.

Once the highest doses of medications used in Arm A and Arm B are determined, then a phase II study will open for children or young adults with Ewing sarcoma that has recurred or is refractory following treatment received after the initial diagnosis. The trial will test the same 2 combinations of therapy in Arm A and Arm B. In the phase II, a participant with Ewing sarcoma will be randomly assigned to receive the treatment given on either Arm A or Arm B.


Condition or disease Intervention/treatment Phase
Recurrent Solid Tumor Recurrent Ewing Sarcoma Recurrent Hepatoblastoma Recurrent Malignant Germ Cell Tumor Recurrent Malignant Solid Neoplasm Recurrent Neuroblastoma Recurrent Osteosarcoma Recurrent Peripheral Primitive Neuroectodermal Tumor Recurrent Rhabdoid Tumor Recurrent Rhabdomyosarcoma Recurrent Soft Tissue Sarcoma Recurrent Wilms Tumor Refractory Ewing Sarcoma Refractory Hepatoblastoma Refractory Malignant Germ Cell Tumor Refractory Malignant Solid Neoplasm Refractory Neuroblastoma Refractory Osteosarcoma Refractory Peripheral Primitive Neuroectodermal Tumor Refractory Rhabdoid Tumor Refractory Rhabdomyosarcoma Refractory Soft Tissue Sarcoma Drug: Onivyde Drug: Talazoparib Drug: Temozolomide Phase 1 Phase 2

Show Show detailed description

Layout table for study information
Study Type : Interventional  (Clinical Trial)
Estimated Enrollment : 160 participants
Allocation: Randomized
Intervention Model: Parallel Assignment
Masking: None (Open Label)
Primary Purpose: Treatment
Official Title: A Randomized Phase I/II Study of Talazoparib or Temozolomide in Combination With Onivyde in Children With Recurrent Solid Malignancies and Ewing Sarcoma
Actual Study Start Date : June 9, 2021
Estimated Primary Completion Date : December 31, 2025
Estimated Study Completion Date : December 31, 2025


Arm Intervention/treatment
Active Comparator: (Arm A) ONI plus TAL
The phase I/II study will evaluate a treatment regimen; nanoliposomal irinotecan (nal-IRN, Onivyde) plus talazoparib (TAL)
Drug: Onivyde
Given intravenous on Days 1 and 8
Other Name: Nanoliposomal irinotecan

Drug: Talazoparib
Given orally twice on Day 1 (daily maximum is 1000mcg/day), then daily on Days 2-6
Other Names:
  • BMN 673
  • Talzenna

Active Comparator: (Arm B) ONI plus TMZ
The phase I/II study will evaluate a treatment regimen; Onivyde (ONI) plus temozolomide (TMZ)
Drug: Onivyde
Given intravenous on Days 1 and 8
Other Name: Nanoliposomal irinotecan

Drug: Temozolomide
Given once a day on Days 1-5.
Other Name: Temodar, Temodal




Primary Outcome Measures :
  1. Phase I:To determine the recommended phase 2 doses (RP2Ds) of Onivyde combined with talazoparib (Arm A) and Onivyde combined with temozolomide (Arm B) administered to children, adolescents and young adults with refractory or recurrent solid malignancies. [ Time Frame: approximately 21 days ]
    First cycle dose limiting toxicity (DLTs) will be used to determine the RP2Ds of the two treatment arms. The DLT is defined as any of the following events that are possibly, probably or definitely attributable to protocol therapy.

  2. Phase II:To estimate the progression-free survival (PFS) of Onivyde plus talazoparib and Onivyde plus temozolomide in patients with refractory or recurrent Ewing sarcoma. [ Time Frame: for five years following the date the last patient was randomized ]
    The study is designed to compare the progression-free survival (PFS) of Onivyde plus talazoparib and Onivyde plus temozolomide in patients with refractory or recurrent Ewing sarcoma. PFS is defined as the time from randomization to disease progression or death, whichever is earlier.


Secondary Outcome Measures :
  1. Phase I:To characterize the safety profile of the treatment regimens, Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of treatment (up to 24 months after enrollment of last participant)] ]
    Safety and tolerability will consist of monitoring and recording all AEs and SAEs as characterized by type, frequency, severity, timing, seriousness and the relationship to the study therapy. These will be reported out qualitatively.

  2. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: maximum plasma concentration (Cmax) on Day 1 and Day 6; for Onivyde: maximum plasma concentration (Cmax) on Day 1 .

  3. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: Tmax on Day 1 and Day 6; for Onivyde: Tmax on Day 1 .

  4. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For both talazoparib and Onivyde: clearance (CL) in Cycle 1.

  5. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: area under the plasma concentration time curve (AUCtau (AUC24)) in Cycle 1.

  6. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: Ctrough (predose) in Cycle 1.

  7. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For Onivyde: area under the plasma concentration time curve (AUCinf) in Cycle 1.

  8. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For Onivyde: half-life (t½) on Day 1.

  9. To characterize the plasma pharmacokinetics of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided For Onivyde: Vd on Day 1.

  10. To estimate the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At time of randomization until evaluation of respective endpoints, up to 4 months after enrollment of last participant ]
    Descriptive statistics of objective response rate (ORR) at cycle 4 will be provided.

  11. To estimate the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At time of randomization until evaluation of respective endpoints, up to 4 months after enrollment of last participant ]
    Descriptive statistics of disease control rate (DCR) at cycle 4 will be provided.

  12. To estimate the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At time of randomization until evaluation of respective endpoints up to 4 months after enrollment of last participant ]
    Descriptive statistics of duration of response (DoR) will be provided.

  13. To estimate the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At time of randomization until evaluation of respective endpoints, up to 2 years after enrollment of last participant ]
    Descriptive statistics event-free survival (EFS) will be provided.

  14. To estimate the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At time of randomization until evaluation of respective endpoints, up to 2 years after enrollment of last participant ]
    Descriptive statistics of overall survival (OS) will be provided.

  15. Phase II: To describe the toxicity of the treatment regimens. [ Time Frame: At the end of treatment (up to 24 months after enrollment of last participant) ]
    Safety and tolerability will consist of monitoring and recording all AEs and SAEs as characterized by type, frequency, severity, timing, seriousness and the relationship to the study therapy. These will be reported out qualitatively.

  16. To describe the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of treatment, up to 24 months after enrollment of last participant ]
    Descriptive statistics of objective response rate (ORR) at cycle 4 will be provided.

  17. To describe the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of treatment, up to 24 months after enrollment of last participant ]
    Descriptive statistics of disease control rate (DCR) at cycle 4 will be provided.

  18. To describe the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of treatment, up to 24 months after enrollment of last participant ]
    Descriptive statistics of duration of response (DoR) will be provided.

  19. To describe the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of treatment, up to 5 years after randomization of last participant ]
    Descriptive statistics of event-free survival (EFS) will be provided.

  20. To describe the antitumor activity of Onivyde plus talazoparib (Arm A) and Onivyde plus temozolomide (Arm B) [ Time Frame: At the end of treatment, up to 5 years after randomization of last participant ]
    Descriptive statistics of overall survival (OS) will be provided.

  21. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: maximum plasma concentration (Cmax) on Day 1 and Day 6; for Onivyde: maximum plasma concentration (Cmax) on Day 1 .

  22. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: Tmax on Day 1 and Day 6; for Onivyde: Tmax on Day 1 .

  23. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For both talazoparib and Onivyde: clearance (CL) in Cycle 1.

  24. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: area under the plasma concentration time curve (AUCtau (AUC24)) in Cycle 1.

  25. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For talazoparib: Ctrough (predose) in Cycle 1.

  26. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma [ Time Frame: At the end of cycle 1 (approximately 21 days after last participant enrollment) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For Onivyde: area under the plasma concentration time curve (AUCinf) in Cycle 1.

  27. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided. For Onivyde: half-life (t½) on Day 1.

  28. To characterize the plasma pharmacokinetics Onivyde and talazoparib in children, adolescents and young adults with refractory or recurrent Ewing sarcoma. [ Time Frame: At the end of Cycle 1 (each cycle is 21 days) ]
    Descriptive statistics of plasma drug concentrations and pharmacokinetic parameters will be provided For Onivyde: Vd on Day 1.



Information from the National Library of Medicine

Choosing to participate in a study is an important personal decision. Talk with your doctor and family members or friends about deciding to join a study. To learn more about this study, you or your doctor may contact the study research staff using the contacts provided below. For general information, Learn About Clinical Studies.


Layout table for eligibility information
Ages Eligible for Study:   12 Months to 30 Years   (Child, Adult)
Sexes Eligible for Study:   All
Accepts Healthy Volunteers:   No
Criteria

Inclusion Criteria

Patients must be > 12 months and < 30 years at the time of enrollment on study.

Phase I

  • Patients with refractory or recurrent non-central nervous system (CNS) solid tumors not amenable to curative treatment are eligible. Patients must have had histologic verification of malignancy at original diagnosis or at the time of relapse. Patients eligible for the expansion cohort, A2, will include non-ES patients with refractory or recurrent non-CNS solid tumors with a deleterious alteration in germline or somatic genes involved in HR repair and DSBs signaling, germline or somatic assessed by prior comprehensive sequencing performed in a CLIA-approved (or equivalent) facility.

Phase II

  • Patients with refractory or recurrent Ewing sarcoma (during or after completion of first-line therapy). Refractory disease is defined as progression during first line treatment or within 12 weeks of completion of first line treatment. Recurrent disease includes patients who received first line treatment and experienced disease progression at any time point >12 weeks from the completion of first line therapy.
  • Patients must have a histologic diagnosis of Ewing sarcoma with EWSR1- FLI1 translocation or other EWS rearrangement at the time of initial diagnosis. Repeat biopsy at the time of disease recurrence is strongly encouraged but it is not required/mandated for enrollment.

Disease status

  • Patients must have either measurable or evaluable disease (see Section 7.0 for definitions). Measurable disease includes soft tissue disease evaluable by cross-sectional imaging (RECIST). Patients with bone disease without a measurable soft tissue component or bone marrow disease only are eligible for the phase 1 and phase 2 study but will not be included in the OR endpoint.
  • Performance level: Karnofsky > 50% for patients > 16 years of age and Lansky > 50% for patients < 16 years of age. Patients who are unable to walk because of paralysis, but who are up in a wheelchair, will be considered ambulatory for the purpose of assessing the performance score.

Prior therapy

Phase I Patients who have received prior therapy with an irinotecan-based or temozolomide-based regimen are eligible. Patients who have received prior therapy with a PARP inhibitor other than talazoparib are eligible.

Phase II

  • Patients should have received first line therapy and developed either refractory or recurrent disease (first relapse).
  • Organ function: Must have adequate organ and bone marrow function as defined by the following parameters:
  • Patients with solid tumors not metastatic to bone marrow:
  • Peripheral absolute neutrophil count (ANC) >1,000/mm3 (1x109/L)
  • Platelet count > 75,000/mm3 (75x109/L) (no transfusion within 7 days of enrollment)
  • Hemoglobin > 9 g/dL (with or without support)

In the phase I study, patients with solid tumors metastatic to bone marrow or with bone marrow hypocellularity defined as <30% cellularity in at least one bone marrow site will be eligible for study, but they will not be evaluable for hematologic toxicity. These patients must not be refractory to red cell or platelet transfusions. At least 2 of every cohort of 3 patients (in the phase I study) must be evaluable for hematologic toxicity. If dose limiting hematologic toxicity is observed at any dose level, all subsequent patients enrolled at that dose level must be evaluable for hematologic toxicity.

  • Adequate renal function defined as: Creatinine clearance or radioisotope GFR > 60ml/min/1.73m2 or a serum creatinine maximum based on age/sex: age 6months to <1 year, creatinine 0.4; 1 to < 2 years, creatinine 0.6; 2 < 6 years, creatinine 0.8; 6 < 10 years, creatinine 1; 10 to <13 years, creatinine 1.2; 13 to < 16 years creatinine 1.5 (males) or 1.4 (females); > 16 years, creatinine 1.7 (males) 1.4 (females)
  • Adequate liver function defined as: normal liver function as defined by SGPT (ALT) concentration <5x the institutional ULN, a total bilirubin concentration <2x the institutional ULN for age, and serum albumin > 2g/dL.
  • Adequate pulmonary function defined as no evidence of dyspnea at rest and a pulse oximetry > 94% if there is a clinical indication for determination. Pulmonary function tests are not required.
  • Patients must have fully recovered from the acute toxic effects of chemotherapy, immunotherapy, surgery, or radiotherapy prior to entering this study:
  • Myelosuppressive chemotherapy: Patient has not received myelosuppressive chemotherapy within 3 weeks of enrollment onto this study (8 weeks if received prior myeloablative therapy).
  • Hematopoietic growth factors: At least 7 days must have elapsed since the completion of therapy with a growth factor. At least 14 days must have elapsed after receiving pegfilgrastim.
  • Biologic (anti-neoplastic agent): At least 7 days must have elapsed since completion of therapy with a biologic agent. For agents that have known adverse events occurring beyond 7 days after administration, this period prior to enrollment must be extended beyond the time during which adverse events are known to occur.
  • Monoclonal antibodies: At least 3 half-lives must have elapsed since prior therapy that included a monoclonal antibody or 28 days have elapsed since last dose of the monoclonal antibody with complete resolution of symptoms related to treatment.
  • Radiotherapy: At least 2 weeks must have elapsed since any irradiation; at least 6 weeks must have elapsed since craniospinal RT, 131I-mIBG therapy or substantial bone marrow irradiation (e.g., >50% pelvis irradiation).
  • Female participant who is post-menarchal must have a negative urine or serum pregnancy test and must be willing to have additional serum and urine pregnancy tests during the study.
  • Female or male participant of reproductive potential must agree to use effective contraceptive methods at screening and throughout duration of study treatment.

Exclusion Criteria

Pregnant or breastfeeding

  • Pregnant or breast-feeding women will not be entered on this study. Pregnancy tests must be obtained in girls who are post-menarchal. Males or females of reproductive potential may not participate unless they have agreed to use two methods of birth control: a medically accepted barrier of contraceptive method (e.g., male or female condom) and a second method of birth control during protocol therapy. Two highly effective methods of contraception are required for female patients during treatment and for at least 7 months after completing therapy. Male patients with female partners of reproductive potential and/or pregnant partners are advised to use two highly effective methods of contraception during treatment and for at least 4 months after the final dose.
  • Male and female participants must agree not to donate sperm or eggs, respectively, after the first dose of study drug through 105 days and 45 days after the last dose of study drug. Females considered not of childbearing potential include those who are surgically sterile (bilateral salpingectomy, bilateral oophorectomy, or hysterectomy).

Information from the National Library of Medicine

To learn more about this study, you or your doctor may contact the study research staff using the contact information provided by the sponsor.

Please refer to this study by its ClinicalTrials.gov identifier (NCT number): NCT04901702


Contacts
Layout table for location contacts
Contact: Sara Federico, MD 866-278-5833 referralinfo@stjude.org

Locations
Layout table for location information
United States, California
Lucille Packard Children's Hospital Stanford Not yet recruiting
Palo Alto, California, United States, 94304
Contact: Allison Pribnow, MD, MPH    650-723-5535    apribnow@stanford.edu   
Principal Investigator: Allison Pribnow, MD, MPH         
United States, Colorado
Children's Hospital Colorado Not yet recruiting
Aurora, Colorado, United States, 80045
Contact: Carrye Cost, MD    720-777-6775    Carrye.Cost@childrenscolorado.org   
Principal Investigator: Carrye Cost, MD         
United States, District of Columbia
Children's National Medical Center Recruiting
Washington, District of Columbia, United States, 20010
Contact: AeRang Kim, PhD    202-476-5685    AeKim@childrenational.org   
Principal Investigator: AeRang Kim, PhD         
Sub-Investigator: Jeffrey Dome, MD,PhD         
United States, Georgia
Children's Healthcare of Atlanta/Emory University School of Medicine Not yet recruiting
Atlanta, Georgia, United States, 30322
Contact: Thomas Cash, MD, MSc    404-785-6288    thomas.cash@choa.org   
Principal Investigator: Thomas Cash, MD, MSc         
United States, Minnesota
Children's Hospital and Clinics of Minn Recruiting
Minneapolis, Minnesota, United States, 55404
Principal Investigator: Kris Ann Schultz, MD         
United States, Tennessee
St. Jude Children's Research Hospital Recruiting
Memphis, Tennessee, United States, 38105
Contact: Sara M Federico, MD    866-278-5833    referralinfo@stjude.org   
Principal Investigator: Sara M Federico, MD         
United States, Texas
Texas Children's Hospital/ Baylor College of Medicine Recruiting
Houston, Texas, United States, 77030
Contact: Jennifer Foster, MD, MPH    832-824-1000    jhfoster@texaschildrens.org   
Principal Investigator: Jennifer Foster, MD, MPH         
Canada, Ontario
The Hospital for Sick Children Recruiting
Toronto, Ontario, Canada
Contact: Daniel Morgenstern, MB, PhD       daniel.morgenstern@sickkids.ca   
Principal Investigator: Daniel Morgenstern, MB, PhD         
Sub-Investigator: Sarah Cohen-Gogo, MD, PhD         
Canada
CHU Sainte-Justine Recruiting
Montréal, Canada
Contact: Henrique Bittencourt, MD,PhD       Henrique.bittencourt.hs@ssss.gouv.qc.ca   
Principal Investigator: Henrique Bittencourt, MD,PhD         
BC Children's Hospital Research Institute Not yet recruiting
Vancouver, Canada
Contact: Rebecca Deyell, MD, FRCPC       RDeyell@cw.bc.ca   
Principal Investigator: Rebecca Deyell, MD, FRCPC         
Sponsors and Collaborators
St. Jude Children's Research Hospital
Pfizer
Ipsen
Investigators
Layout table for investigator information
Principal Investigator: Sara Federico, MD St. Jude Children's Research Hospital
Additional Information:
Layout table for additonal information
Responsible Party: St. Jude Children's Research Hospital
ClinicalTrials.gov Identifier: NCT04901702    
Other Study ID Numbers: ONITT
NCI-2021-02843 ( Other Identifier: NCI )
First Posted: May 25, 2021    Key Record Dates
Last Update Posted: December 12, 2023
Last Verified: November 2023
Individual Participant Data (IPD) Sharing Statement:
Plan to Share IPD: Yes
Plan Description: Individual participant de-identified datasets containing the variables analyzed in the published article will be made available (related to the study primary or secondary objectives contained in the publication). Supporting documents such as the protocol, statistical analyses plan, and informed consent are available through the CTG website for the specific study. Data used to generate the published article will be made available at the time of article publication. Investigators who seek access to individual level de-identified data will contact the computing team in the Department of Biostatistics (ClinTrialDataRequest@stjude.org) who will respond to the data request.
Supporting Materials: Study Protocol
Statistical Analysis Plan (SAP)
Informed Consent Form (ICF)
Time Frame: Data will be made available at the time of article publication.
Access Criteria: Data will be provided to researchers following a formal request with the following information: full name of requestor, affiliation, data set requested, and timing of when data is needed. As an informational point, the lead statistician and study principal investigator will be informed that primary results datasets have been requested.

Layout table for additional information
Studies a U.S. FDA-regulated Drug Product: Yes
Studies a U.S. FDA-regulated Device Product: No
Keywords provided by St. Jude Children's Research Hospital:
Pediatric Cancer
Childhood Cancer
Ewing Sarcoma
PARP inhibitor
Solid tumor
Irinotecan
Onivyde
Talazoparib
Additional relevant MeSH terms:
Layout table for MeSH terms
Neoplasms
Sarcoma
Neuroblastoma
Neoplasms, Germ Cell and Embryonal
Osteosarcoma
Rhabdomyosarcoma
Sarcoma, Ewing
Neuroectodermal Tumors
Neuroectodermal Tumors, Primitive
Wilms Tumor
Neuroectodermal Tumors, Primitive, Peripheral
Rhabdoid Tumor
Hepatoblastoma
Recurrence
Disease Attributes
Pathologic Processes
Neoplasms, Connective and Soft Tissue
Neoplasms by Histologic Type
Neoplasms, Neuroepithelial
Neoplasms, Glandular and Epithelial
Neoplasms, Nerve Tissue
Neoplasms, Bone Tissue
Neoplasms, Connective Tissue
Myosarcoma
Neoplasms, Muscle Tissue
Neoplasms, Complex and Mixed
Kidney Neoplasms
Urologic Neoplasms
Urogenital Neoplasms
Neoplasms by Site