Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT01962103
Other study ID # ABI-007-PST-001
Secondary ID
Status Completed
Phase Phase 1/Phase 2
First received
Last updated
Start date December 4, 2013
Est. completion date November 6, 2018

Study information

Verified date December 2019
Source Celgene
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

The purpose of this study is to find the safe dose of nab-paclitaxel in children with solid tumors, and to see if it works to treat these solid tumors in children and young adults (in Phase 1 ≤ 18 years old and in Phase 2 ≤ 24 years old). After the final dose has been chosen, patients will be enrolled according to the specific solid tumor type, (neuroblastoma, rhabdomyosarcoma, or Ewing's sarcoma), to see how nab-paclitaxel works in treating these tumors.


Description:

ABI-007-PST-001 is a Phase 1/2, multicenter, open-label, dose-finding study to assess the safety , tolerability, and preliminary efficacy of weekly nab-paclitaxel in pediatric patients with recurrent or refractory solid tumors (excluding brain tumors). The Phase 1 portion of the study, with a dose escalation design, ended and the recommended Phase 2 dose (RP2D) was determined as 240 mg/m^2 intravenously (IV) in patients weighing > 10 kg and 11.5 mg/kg in patients weighing ≤ 10 kg, on Days 1, 8 and 15 of a 28-day cycle. The Phase 2 portion of the study will enroll additional patients at the RP2D into 1 of 3 solid tumor groups [neuroblastomas, rhabdomyosarcomas, Ewing's sarcomas]. Both phases of the study are open-label and conducted at multiple centers.

The Phase 2 is using a Simon 2-stage design to monitor patient enrollment for each group separately. The rhabdomyosarcoma group, neuroblastoma or Ewing's sarcoma groups did not reach the expected number of 2 responders out of 14 efficacy eligible patients. Consequently, the groups were stopped.


Recruitment information / eligibility

Status Completed
Enrollment 107
Est. completion date November 6, 2018
Est. primary completion date December 5, 2017
Accepts healthy volunteers No
Gender All
Age group 6 Months to 24 Years
Eligibility Inclusion Criteria:

- Patients must meet all of the following criteria to be enrolled in the study:

1. Patient has a confirmed solid tumor diagnosis according to the

following:

1. Phase 1: patient has a recurrent or refractory solid tumor that has

progressed or did not respond to standard therapy, or for which no

standard anticancer therapy exists

2. Phase 2: patient has radiologically documented measurable disease by Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 (for neuroblastoma, evaluable disease by 123^I-metaiodobenzylguanidine [MIBG]/Curie score is also acceptable) in 1 of the following tumor types and has failed up to 3 lines of treatment: Group 1: neuroblastoma, Group 2: rhabdomyosarcoma; Group 3: Ewing's sarcoma.

2. The patient has a Lansky/Karnofsky performance status score of = 70%.

3. The patient has adequate serum chemistry levels, evidenced by the

following laboratory values

1. aspartate aminotransferase (AST)/serum glutamic-oxaloacetric

transaminase (SGOT), alanine aminotransferase (ALT)/serum glutamic

pyruvate transaminase (SGPT) = 2.5 × upper limit of normal range (ULN)

2. Total bilirubin = 1.5 × ULN

3. Creatinine = 1.5 × ULN

4. The patient has adequate bone marrow function, evidenced by the

following:

1. Absolute neutrophil count = 1.0 × 10^9 cells/L

2. Platelets = 80 × 10^9 cells/L (transfusion independent, defined as not

receiving platelet transfusions within 7 days prior to laboratory sample). In the phase 2 portion, for patients with known bone marrow involvement, platelets = 50 × 10^9 cells/L

3. Hemoglobin = 8 g/dL (transfusion is permitted to fulfill this criterion).

5. The patient (when applicable) or patient's parent(s) or legal guardian(s)

understand(s) and voluntarily signed an informed consent document prior

to any study-related assessments/procedures being conducted. Where

locally applicable, the patient also understands and voluntarily provides

his/her assent prior to any study-related assessments/procedures being

conducted.

6. Male patients of childbearing potential must use a condom during

sexual intercourse and shall not father a child during the study and for 6 months after the last dose of study medication.

7. Female patients of childbearing potential [defined as all female

patients = 12 years old or who have reached menarche, whichever occurs

first] must have both of the following:

a. Agree to the use of two physician-approved contraceptive methods

simultaneously or practice complete abstinence while on study medication or for a longer period if required by regulations.

i. True abstinence: When this is in line with the preferred and usual

lifestyle of the patient. Periodic abstinence (e.g., calendar, ovulation,

symptothermal, postovulation methods) and withdrawal are not

acceptable methods of contraception.

ii. Acceptable contraceptive methods include: oral, injectable, or

implantable hormonal contraceptive; tubal ligation; intra-uterine device;

barrier contraceptive with spermicide; or vasectomized partner) including

at least one barrier method.

b. Have negative serum pregnancy test result at screening confirmed by

negative urine pregnancy dipstick within 72 hours prior to first dose of

investigational product (if serum test occurred > 72 hours from first

dose); pregnancy test with sensitivity of at least 25 mIU/mL.

Exclusion Criteria:

- The presence of any of the following will exclude a patient from enrollment:

1. The patient has a primary brain tumor(s) or brain metastasis (unless metastasis is treated and stable for > 28 days). In patients who are symptomatic, a brain scan is required to exclude metastasis.

2. The patient has received therapeutic dose chemotherapy or radiotherapy = 21 days prior to start of investigational product.

3. The patient has received maintenance dose chemotherapy (e.g., low dose cyclophosphamide) = 7 days from the first dose of investigational product.

4. The patient has received any investigational therapy = 28 days prior to start of investigational product. Investigational therapy is defined as any medicinal product that is not approved in the country of treatment for any indication, adult or pediatric.

5. The patient has received any biological therapy = 7 days prior to the start of investigational product, or monoclonal antibody = 3 half-lives or 28 days, whichever is shorter, prior to the first dose of investigational product.

6. The patient has received any hematopoietic stem cell transplantation (HSCT) = 3 months prior to start of investigational product.

7. The patient has received allogeneic hematopoietic stem cell transplantation (HSCT) = 3 months or autologous HSCT = 21 days prior to start of investigational product.

8. The patient has not recovered from the acute toxic effects of prior chemotherapy, radiation, or major surgery/significant trauma.

9. The patient has had minor surgery = 7 days from the start of study treatment (excluding the placement of central/peripheral lines, skin biopsy).

10. The patient has a known history of stroke, myocardial infarction, peripheral vascular disease, or recent (within 3 months) uncontrolled deep venous thrombosis.

11. The patient has a known history or current diagnosis of human immunodeficiency virus (HIV) infection, regardless of treatment status.

12. The patient has an uncontrolled intercurrent illness including but not limited to ongoing or active infection requiring antibiotic, antifungal, or antiviral therapy, symptomatic heart failure, cardiac arrhythmia, or psychiatric illness/social situations that would limit compliance with study requirements.

13. The patient has any significant medical condition, laboratory abnormality, or psychiatric illness that would prevent the patient from participating in the study.

14. The patient has any condition, including the presence of laboratory abnormalities, that places the patient at unacceptable risk if he/she were to participate in the study.

15. The patient has any condition that confounds the ability to interpret data from the study.

16. The patient or parent(s)/guardian(s) is/are unable to comply with the study visit schedule and other protocol requirements, in the opinion of the investigator.

17. The patient has = Grade 2 peripheral neuropathy by National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) at screening.

Study Design


Related Conditions & MeSH terms

  • Cancer
  • Clinical Oncology
  • Dermatofibrosarcoma
  • Ewing's Sarcoma
  • Ewing's Tumor
  • Fibrosarcoma
  • Histiocytoma
  • Malignant Melanoma
  • Melanoma
  • Neoplasia
  • Neoplasm
  • Neoplasms
  • Neuroblastoma
  • Neuroectodermal Tumors, Primitive, Peripheral
  • Oncology, Medical
  • Osteogenic Sarcoma
  • Osteosarcoma
  • Osteosarcoma Tumor
  • Pediatrics, Osteosarcoma
  • Rhabdomyosarcoma
  • Sarcoma
  • Sarcoma, Ewing
  • Sarcoma, Ewing's
  • Sarcoma, Osteogenic
  • Sarcoma, Soft Tissue
  • Sarcoma, Spindle Cell
  • Sarcomas, Epitheliod
  • Tumors

Intervention

Drug:
nab-paclitaxel
nab-paclitaxel 120-270 mg/m2 IV on Days 1, 8 and 15 of a 28-day cycle
nab-paclitaxel
IV infusion

Locations

Country Name City State
Canada The Hospital for Sick Children Toronto Ontario
France Institute for Pediatric Hematology - Oncology, Leon Berard Cancer Center Lyon
France Hopital d'Enfants, CHU Nancy Nancy
France Institut Curie Paris
France Institut Gustave Roussy Villejuif
Italy Azienda Ospedaliera Universitaria Meyer Firenze
Italy Children's Hospital Largo Genova
Italy Istituto Nazionale Tumori Milan
Italy Clinica di Oncoematologia Padova
Italy Policlinico Agostino Gemelli Rome
Italy l'Azienda Ospedaliera Regina Margherita - Sant Anna Torino
Spain Hospital Sant Joan de Deu Barcelona
Spain Hospital Universitario Vall D Hebron Barcelona
Spain Spanish National Cancer Research Centre Madrid
Spain Hospital Universitario Virgen Del Rocio Sevilla
Spain Unidad de Oncologia Pediatrica, Hospital Universitario la Fe Valencia
Switzerland Universitäts-Kinderklinik Zurich
United Kingdom Royal Marsden Hospital Sutton
United States Columbia University Medical Center New York New York
United States Phoenix Childrens Hospital Phoenix Arizona

Sponsors (1)

Lead Sponsor Collaborator
Celgene

Countries where clinical trial is conducted

United States,  Canada,  France,  Italy,  Spain,  Switzerland,  United Kingdom, 

References & Publications (1)

Moreno L, Casanova M, Chisholm JC, Berlanga P, Chastagner PB, Baruchel S, Amoroso L, Gallego Melcón S, Gerber NU, Bisogno G, Fagioli F, Geoerger B, Glade Bender JL, Aerts I, Bergeron C, Hingorani P, Elias I, Simcock M, Ferrara S, Le Bruchec Y, Slepetis R, Chen N, Vassal G. Phase I results of a phase I/II study of weekly nab-paclitaxel in paediatric patients with recurrent/refractory solid tumours: A collaboration with innovative therapies for children with cancer. Eur J Cancer. 2018 Sep;100:27-34. doi: 10.1016/j.ejca.2018.05.002. Epub 2018 Jun 21. — View Citation

Outcome

Type Measure Description Time frame Safety issue
Primary Phase 1: Number of Participants With Dose Limiting Toxicities (DLTs) A DLT was defined as investigational product (IP)-related adverse events occurring during the DLT assessment period that led to treatment discontinuation or met one of the following criteria: - Common Terminology Criteria for Adverse Events (CTCAE) Grade (Gr) 3 or 4 nonhematologic toxicity (excluding transient transaminitis) - CTCAE Gr 3 or 4 nausea or vomiting that persisted > 5 days despite maximal anti-emetic treatment - CTCAE Gr 4 thrombocytopenia or anemia that persisted > 7 days or required transfusion > 7 days - CTCAE Gr 3 thrombocytopenia with bleeding - CTCAE Gr 4 uncomplicated neutropenia lasting > 7 days - Febrile neutropenia with confirmed bacterial infection - CTCAE Gr 3 hematologic toxicity requiring treatment (tx) delay > 21 days. Use of "..." in the table rows signifies the continuation of row title per the above list. DLT assessment period: For participants > 10 kg: the first 28-day cycle including Cycle 2 Day 1 predose evaluations; for participants = 10 kg: the first two 28-day cycles including Cycle 3 Day 1 predose evaluations
Primary Phase 1: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) An adverse event (AE) was defined as any noxious, unintended, or untoward medical occurrence that may appear or worsen in a participant during the course of a study. A serious AE (SAE) is any AE occurring at any dose that: results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. TEAEs were defined as AEs that began or worsened in severity on or after the date of the first dose of study drug and within 28 days of the date of the last dose of study drug. The severity of an AE was graded according to the CTCAE, Version 4.0. Median treatment duration in Phase 1 was 7.0 weeks, with minimum and maximum duration of 1 and 49 weeks, respectively. Participants were followed for 28 days after discontinuing treatment for safety and monitoring of AEs.
Primary Phase 2: Overall Response Rate (ORR) Overall response rate was defined as the percentage of participants who achieved a complete response (CR; disappearance of all target lesions) or partial response (PR; at least a 30% decrease in the sum of diameters of target lesions) confirmed no less than 4 weeks after the criteria for response were first met using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 guidelines. (For Phase 2 neuroblastoma participants who had both RECIST and Curie Score tumor evaluations, both tumor response results were considered and an overall response was derived.) Confidence interval was obtained using the Clopper-Pearson method. Median treatment duration in Phase 2 per group: Ewings Sarcoma = 14 weeks (3-31), Neuroblastoma = 7 weeks (3-23), Rhabdomyosarcoma = 5 weeks (1-13).
Secondary Phase 1: ORR Overall response rate was defined as the percentage of participants who achieved a complete response (CR; disappearance of all target lesions) or partial response (PR: at least a 30% decrease in the sum of diameters of target lesions) confirmed no less than 4 weeks after the criteria for response were first met) using RECIST version 1.1 guidelines over the total number of participants available for the analysis. Confidence interval was obtained using the Clopper-Pearson method. Median treatment duration in Phase 1 was 7.0 weeks, with minimum and maximum duration of 1 and 49 weeks, respectively.
Secondary Phase 1: Maximum Observed Concentration of Paclitaxel in Blood Plasma (Cmax) Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: Cmax - Dose-Normalized Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: Area Under the Plasma Concentration-Time Curve (AUC) Measurements include: AUC from time zero to the last measurable concentration (AUCt), AUC from time zero to 24 hours (AUC24), and AUC from time zero to infinity (AUCinf). Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: AUC - Dose-Normalized Measurements include: AUC24 and AUCinf. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: Clearance (CL) Measurement of renal clearance from the body. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: CL - Body Surface Area (BSA)-Normalized Measurement of renal clearance from the body. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: Volume of Distribution (Vss) Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1: Vss - BSA-Normalized Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants <6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Volume of Distribution of the Central Compartment (V1) Population PK analysis was performed using nonlinear mixed effect modeling. The estimated allometric function for V1 was 0.888. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Maximum Elimination Rate From the Central Compartment (VMEL) Population PK analysis was performed using nonlinear mixed effect modeling. The estimated allometric function for VMEL was 1.12. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Concentration in the Central Compartment at 50% of VMEL (KMEL) Population PK analysis was performed using nonlinear mixed effect modeling. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Intercompartmental CL Between the Central Compartment and the First Peripheral Compartment (Q2) Population PK analysis was performed using nonlinear mixed effect modeling. The estimated allometric function for Q2 was 1.12. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Intercompartmental CL Between the Central Compartment and the Second Peripheral Compartment (Q3) Population PK analysis was performed using nonlinear mixed effect modeling. The estimated allometric function for Q3 was 1.12. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Volume of Distribution of the First Peripheral Compartment (V2) Population PK analysis was performed using nonlinear mixed effect modeling. The estimated allometric function for V2 was 0.888. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 1 and 2 Population PK: Volume of Distribution of the Second Peripheral Compartment (V3) Population PK analysis was performed using nonlinear mixed effect modeling. The estimated allometric function for V3 was 0.888. Cycle 1 Day 1 (Participants = 6 years: 1-2 minutes prior to the end of infusion [EOI], and 15 minutes, 1, 3, 5, 8, 24, 48, and 72 hours after the EOI. Participants < 6 years: 1-2 minutes prior to the EOI, and 15 minutes, 3, 5, and 24 hours after the EOI.)
Secondary Phase 2: Duration of Response (DOR) Duration of response was defined as the time from the date of the first response (CR/PR, using RECIST version 1.1 guidelines) to disease progression for participants with a confirmed CR or PR. Participants who did not have disease progression or had not died were censored at the time of their last disease assessment or at time of start of new anticancer therapy, whichever occurred first. (For Phase 2 neuroblastoma patients who had both RECIST version 1.1 and Curie Score tumor evaluations, both tumor responses results were considered and an overall response was derived.) Median treatment duration in Phase 2 per group: Ewings Sarcoma = 14 weeks (3-31), Neuroblastoma = 7 weeks (3-23), Rhabdomyosarcoma = 5 weeks (1-13). Participants were followed until disease progression (if applicable) up to a maximum of 100.3 weeks.
Secondary Phase 2: Disease Control Rate (DCR) Disease control rate was defined as the percentage of participants who achieved either a stable disease maintained for = 16 weeks or confirmed CR (confirmed no less than 4 weeks after criteria for response were first met) or confirmed PR (confirmed no less than 4 weeks after criteria for response were first met) over the total number of participants available for the analysis. Confidence interval was obtained using the Clopper-Pearson method. Median treatment duration in Phase 2 per group: Ewings Sarcoma = 14 weeks (3-31), Neuroblastoma = 7 weeks (3-23), Rhabdomyosarcoma = 5 weeks (1-13).
Secondary Phase 2: Progression-Free Survival (PFS) PFS was defined as the time from the first dose date to the start of disease progression or participant death (any cause), whichever occurred first. Disease progression was classed as either a disease progression observed as a response assessment, or a disease progression or symptomatic deterioration at treatment/study discontinuation. Participants who did not have disease progression or had not died were censored at the last known time that the participant was progression free. Disease progression was considered according to RECIST version 1.1 for Phase 2 Ewing's sarcoma and rhabdomyosarcoma participants. (For Phase 2 neuroblastoma participants who had both RECIST 1.1 and Curie score tumor evaluations, both tumor responses results were considered and an overall response was derived.) Median PFS time was estimated through Kaplan-Meier methods. 95% confidence interval about the median time to PFS event was obtained using Greenwood's method. Median treatment duration in Phase 2 per group: Ewings Sarcoma = 14 weeks (3-31), Neuroblastoma = 7 weeks (3-23), Rhabdomyosarcoma = 5 weeks (1-13). Participants were followed until disease progression (if applicable) up to a maximum of 100.3 weeks.
Secondary Phase 2: Kaplan-Meier Estimate of Overall Survival Rate at 1 Year Overall survival was defined as the time from the first dose date to date of death (any cause). Participants who were alive were censored at the last known time that the participant was alive. 1 year
Secondary Phase 2: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) An AE was defined as any noxious, unintended, or untoward medical occurrence that may appear or worsen in a participant during the course of a study. A SAE is any AE occurring at any dose that: results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. TEAEs were defined as AEs that began or worsened in severity on or after the date of the first dose of study drug and within 28 days of the date of the last dose of study drug. The severity of the AEs was graded according to the Common Terminology Criteria for Adverse Events, Version 4.0. Participants were followed for 28 days after discontinuing treatment for safety and monitoring of AEs. Median treatment duration in Phase 2 per group: Ewings Sarcoma = 14 weeks (3-31), Neuroblastoma = 7 weeks (3-23), Rhabdomyosarcoma = 5 weeks (1-13). Participants were followed for 28 days after discontinuing treatment for safety and monitoring of AEs.
See also
  Status Clinical Trial Phase
Recruiting NCT05346796 - Survivorship Plan HEalth REcord (SPHERE) Implementation Trial N/A
Recruiting NCT05094804 - A Study of OR2805, a Monoclonal Antibody Targeting CD163, Alone and in Combination With Anticancer Agents Phase 1/Phase 2
Completed NCT04867850 - Effect of Behavioral Nudges on Serious Illness Conversation Documentation N/A
Enrolling by invitation NCT04086251 - Remote Electronic Patient Monitoring in Oncology Patients N/A
Completed NCT01285037 - A Study of LY2801653 in Advanced Cancer Phase 1
Completed NCT00680992 - Study of Denosumab in Subjects With Giant Cell Tumor of Bone Phase 2
Completed NCT00062842 - Study of Irinotecan on a Weekly Schedule in Children Phase 1
Active, not recruiting NCT04548063 - Consent Forms in Cancer Research: Examining the Effect of Length on Readability N/A
Completed NCT04337203 - Shared Healthcare Actions and Reflections Electronic Systems in Survivorship N/A
Recruiting NCT04349293 - Ex-vivo Evaluation of the Reactivity of the Immune Infiltrate of Cancers to Treatments With Monoclonal Antibodies Targeting the Immunomodulatory Pathways N/A
Terminated NCT02866851 - Feasibility Study of Monitoring by Web-application on Cytopenia Related to Chemotherapy N/A
Active, not recruiting NCT05304988 - Development and Validation of the EFT for Adolescents With Cancer
Completed NCT04448041 - CRANE Feasibility Study: Nutritional Intervention for Patients Undergoing Cancer Surgery in Low- and Middle-Income Countries
Completed NCT00340522 - Childhood Cancer and Plexiform Neurofibroma Tissue Microarray for Molecular Target Screening and Clinical Drug Development
Recruiting NCT04843891 - Evaluation of PET Probe [64]Cu-Macrin in Cardiovascular Disease, Cancer and Sarcoidosis. Phase 1
Active, not recruiting NCT03844048 - An Extension Study of Venetoclax for Subjects Who Have Completed a Prior Venetoclax Clinical Trial Phase 3
Completed NCT03167372 - Pilot Comparison of N-of-1 Trials of Light Therapy N/A
Completed NCT03109041 - Initial Feasibility Study to Treat Resectable Pancreatic Cancer With a Planar LDR Source Phase 1
Terminated NCT01441115 - ECI301 and Radiation for Advanced or Metastatic Cancer Phase 1
Recruiting NCT06206785 - Resting Energy Expenditure in Palliative Cancer Patients