Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT01456039
Other study ID # ROMI-TCL-001
Secondary ID
Status Completed
Phase Phase 1/Phase 2
First received
Last updated
Start date December 1, 2011
Est. completion date December 14, 2018

Study information

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

Clinical Trial Summary

The purpose of the study was to assess efficacy, tolerability, safety and pharmacokinetics of Romidepsin in subjects with progressive or relapsed peripheral T-cell lymphoma


Description:

This is a Phase 1/2, non-randomized, open-label, single-arm trial with two phases. The first phase is a 3 + 3 dose escalation phase to determine a recommended dose for treating patients with Peripheral T-Cell Lymphoma (PTCL) or Cutaneous T-Cell Lymphoma (CTCL) based on the assessment of Dose Limiting Toxicities (DLTs).The second phase will assess efficacy at the recommended dose by measuring objective response [Complete Response (CR), Unconfirmed Complete Response (CR(u)) or Partial Response (PR)] and determining best overall response of each patient. Phase 1 will enroll a maximum of 12 patients and Phase 2 will enroll up to approximately 40 patients


Recruitment information / eligibility

Status Completed
Enrollment 51
Est. completion date December 14, 2018
Est. primary completion date July 28, 2015
Accepts healthy volunteers No
Gender All
Age group 20 Years and older
Eligibility Inclusion Criteria:

Patients must fulfill all of the following criteria to be eligible for study participation and have:

- Histologically confirmed Peripheral T cell Lymphoma (PTCL) Not Otherwise Specified (NOS), Angioimmunoblastic T-cell Lymphoma, enteropathy- type T-cell lymphoma, subcutaneous panniculitis-like T-cell lymphoma, cutaneous T-cell lymphoma (excludes mycosis fungoides or Sezary syndrome) , hepatosplenic T-cell lymphoma, Anaplastic Large cell lymphoma (ALCL) [anaplastic lymphoma kinase-1 (ALK-1) negative], patients with ALK 1 expressing ALCL (ALK-1 positive) who have relapsed disease after Autologous Stem-Cell Transplantation, Transformed mycosis fungoides (MF), or Sézary syndrome (SS);

- Age =20 years;

- Written informed consent;

- Progressive Disease following at least one systemic therapy or refractory to at least one prior systemic therapy;

- Eastern Cooperative Oncology Group (ECOG) performance status of 0-2;

- Sufficient functions of bone marrow or other organs as evidenced by

- Hemoglobin =8.0 g/dL (The value after the 7th day of transfusion)

- Absolute neutrophil count (ANC) =1.0×10^9/L (The value after the 7th day of G-CSF)

- Platelet counts =100 x 10^9/L, or, if bone marrow infiltration is recognized, =75 ×10^9/L

- Total bilirubin (Total-Bil) =2 x upper limit of normal (ULN) (=3.0 x ULN in the presence of demonstrable liver metastasis)

- Aspartate Aminotransferase (AST)/Serum Glutamic-Oxaloacetic Transaminase (SGOT) =2 x Upper Limit Normal (ULN) (=3.0 x ULN in the presence of demonstrable liver metastasis)

- Alanine Aminotransferase (ALT)/Serum Glutamic-Oxaloacetic Transaminase (SGOT) =2 x ULN (=3.0 x ULN in the presence of demonstrable liver metastasis)

- Serum creatinine = 2 x ULN

- Serum potassium = lower limit of normal (LLN) and magnesium

- Patients for whom at least 1 measurable lesion is confirmed in the lesion assessment before enrollment; and

- Negative urine or serum pregnancy test on females of childbearing potential.

Exclusion Criteria: Confirmation should be made before enrollment, and the subjects corresponding to the criteria should not be enrolled.

1. Subjects in whom central nervous lymphoma is recognized during the screening period (If brain metastasis is suspected clinically, CT should be performed.)

2. Subjects undergoing chemotherapy or immunotherapy for the purpose of treatment of the target disease within 22 days before C1D1 (including C1D1) (using antibody drugs only within 3 months before C1D1)

3. Subjects receiving local application of steroids within 15 days before C1D1 (including C1D1) Use of steroids for the purpose other than that of treatment of the target disease should be allowed (Only CTCL subjects)

4. Subjects receiving systemic application of steroids within 22 days before C1D1 (including C1D1) (It is acceptable to continue the use of the steroid for the purpose of treatment of the target disease,which administered in doses = 10mg/day prednisolone or equivalent dose of other glucocorticoid. However increase of steroid dose cannot be allowed during the study period)

5. Subjects undergoing radiation therapy, PUVA therapy or TSEB for the purpose of treatment of the target disease within 22 days before C1D1 (including C1D1)

6. Subjects using other investigational products within 22 days before C1D1 (including C1D1) (using antibody drugs only within 3 months before C1D1)

7. Subjects undergoing blood transfusion and using G-CSF within 8 days before C1D1 (including C1D1)

8. Subjects with the following abnormalities in the cardiac function

1. Congenital QT prolongation syndrome

2. QTc interval >480 msec

3. Myocardial infarction within 6 months before C1D1. Subjects with a history of myocardial infarction between 6 and 12 months prior to C1D1 who are asymptomatic and have had a negative cardiac risk assessment (treadmill stress test, nuclear medicine stress test, or stress echocardiogram) since the event may occur

4. Significant ECG abnormalities including atrio-ventricular (AV) block type II, 3rd degree AV block, or bradycardia (ventricular rate less than 50 beats/min)

5. Symptomatic coronary artery disease (CAD) (e.g., Angina Canadian Class II-IV).

6. An ECG recorded at screening showing significant ST depression (ST depression of =2 mm, measured from isoelectric line to the ST segment at a point 60 msec at the end of the QRS complex). If there is any doubt, the subject should have a stress imaging study and, if abnormal, angiography to define whether or not CAD is present.

7. Congestive heart failure (CHF) that meets New York Heart Association (NYHA) Class II to IV definitions and/or ejection fraction <40% by MUGA scan or <50% by echocardiogram and/or MRI

8. A known history of sustained ventricular tachycardia (VT), ventricular fibrillation (VF), torsade de pointes, or cardiac arrest unless currently addressed with an automatic implantable cardioverter defibrillator (AICD)

9. Hypertrophic cardiomyopathy or restrictive cardiomyopathy from prior treatment or other causes (if there is any doubt, see ejection fraction criteria above)

10. Uncontrolled hypertension, i.e., systolic blood pressure (BP) is greater than or equal to 160 mmHg or diastolic BP is greater than or equal to 95 mmHg; subjects who have a history of hypertension controlled by medication must be on a stable dose (for at least one month)

11. Subjects with cardiac arrhythmia requiring an anti-arrhythmic drug

9. Concomitant use of a drug which may induce significant QT prolongation (refer to 9.2. Prohibited Concomitant Medications and Procedures.)

10. Concomitant use of strong or moderate CYP3A4 inhibitors which include grapefruit juice

11. Concomitant use of CYP3A4 inducers which include St John's Wort (1st step only)

12. Concomitant use of therapeutic warfarin which has a potential drug interaction. Use of a small dose of warfarin or other anticoagulant agent to maintain patency of venous access port and cannulas is permitted.

13. Clinically important active infections

14. Known infection with human immunodeficiency virus (HIV) antibody positive, HBs antigen positive or HCV antibody positive. If negative for HBsAg but HBcAb and/or HBsAb positive status, a HBV DNA test will be performed and if positive the subject will be excluded.

15. Subjects undergoing a wide range of radiation therapy in =30% of the bone marrow (such as all parts of the pelvic area or a half of the spinal cord) in the past. The subjects undergoing systemic radiation (including systemic electron therapy) as previous treatment for ASCT will be excluded.

16. Subjects undergoing a surgery within 15 days before C1D1 (including C1D1); however, even if more than 15 days have passed since the surgery, subjects without evidence of wound healing will be excluded.

17. Subjects who are during recovery process from severe wound or fracture.

18. Subjects with a history of allogeneic stem cell transplantation

19. Patients who are breast-feed during period of the IP administration or within 28 days after the end of the IP administration.

20. Subjects with a history of any other malignant tumor or solid cancer within previous 3 years (excluding basal or squamous cell carcinoma of skin, and in situ carcinoma of the cervix (CIN3) that has been treated curatively)

21. Subject with a history of hematological malignant tumor (other than T-cell lymphoma)

22. Subjects for whom transfusion of red blood cells or platelets is impossible (such as clinical state and religious beliefs)

23. Significant medical or psychiatric situation by which all of the study procedures may not be observed

24. Subjects receiving romidepsin in the past (Other HDAC inhibitors are acceptable)

25. Subjects judged to be inappropriate for this study by the investigator or sub-investigator.

26. Concomitant use of rifampicin.

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Romidepsin
Intravenous dosing for 4 hours on Days 1, 8, and 15 of each 28-day cycle

Locations

Country Name City State
Japan National Cancer Center Hospital Chuo Tokyo
Japan Kyushu University Hospital Fukuoka
Japan Chugoku Central Hospital Fukuyamashi Hiroshima
Japan Tokai University School of Medicine Isehara Kanagawa
Japan National Cancer Center Hospital East Kashiwa Chiba
Japan Kumamoto University Hospital Kumamoto
Japan University hospital, Kyoto prefectural University of Medicine Kyoto
Japan Tohoku University Hospital Miyagi
Japan Nagoya City University Hospital Nagoya Aichi
Japan Nagoya Daini Red Cross Hospital Nagoya Aichi
Japan Kochi Medical School Hospital Nankoku Kochi
Japan Sapporo Hokuyu Hospital Sapporo Hokkaido
Japan Sapporo Medical University Hospital Sapporo Hokkaido
Japan Kinki University Hospital Sayama Osaka
Japan Japanese Red Cross Medical Center Shibuya Tokyo
Japan Ehime University Hospital To-on Ehime
Japan The Cancer Institute Hospital of JFCR Tokyo

Sponsors (1)

Lead Sponsor Collaborator
Celgene

Country where clinical trial is conducted

Japan, 

Outcome

Type Measure Description Time frame Safety issue
Primary Number of Participants With Dose-limiting Toxicity (DLT) in Accordance With National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0 as Determined by the Efficacy and Safety Evaluation Committee (ESEC) DLT was defined as an adverse event (AE) occurring in Cycle 1 in Phase 1 and judged that the causal relationship to the investigational product could not be denied. The severity of all AEs was graded based upon the NCI CTCAE version 3.0. DLTs were defined as: • Grade 4 Hemoglobin <6.5 g/dL • Grade 4 Neutrophil <500/µL continuing for at least 5 days • Febrile neutropenia (Grade 4 neutropenia caused by fever and = 38.5° C for more than 1 hour) • Grade 4 thrombocyte (< 25,000/µL), or thrombocytopenia with hemorrhage requiring platelet transfusion • Nausea, vomiting, or diarrhea at > grade 3 in spite of treatment • Grade 3 ALT (alanine aminotransferase) or AST (aspartate aminotransferase) values continued for 7 days. • Grade 4 ALT or AST • Grade 2 arrhythmia • Grade 4 non-hematological AEs • Other grade 3 non-hematological AEs except transient fatigue, anorexia, hyponatremia, and tumor lysis syndrome • Other AEs leading to discontinuation of administration Up to Day 28; Cycle 1
Primary Percentage of PTCL Participants With an Overall Best Response in Accordance With a Modified International Workshop Response Criteria (IWC) 1999 in Phase 2 Objective disease response in PTCL was defined as patients with a complete response (CR), unconfirmed complete response (CRu) or a partial response (PR) according to modified IWC 1999 criteria and assessed by an independent efficacy reviewer. A CR is >75% decrease in size of maximum 6 largest target within nodal and extranodal lesions, complete disappearance of other nodal and extranodal; total disappearance of clinical disease; disease-related signs and symptoms, normalization of biochemical abnormalities, disappearance of spleen, liver, or kidney enlargement; no bone marrow (BM) involvement, no new sites of disease. CRu: all above criteria fulfilled except for BM involvement is indeterminate. PR: a =50% decrease in size of 6 largest target lesions and no increase other nodal and extranodal; no progression of clinical disease; disease-related signs and symptoms, normalization or biochemical abnormalities, no progression in size of liver, spleen, or kidney; and no new sites of disease Tumor assessments performed every 2 months; median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Participants With Treatment-Emergent Adverse Events (TEAEs) Associated With Romidepsin An adverse event (AE) is any noxious, unintended, or untoward medical occurrence that may appear or worsen during the course of a study. An AE that resulted in any of the outcomes was defined as a serious (SAE): • Death • Life-threatening event • An inpatient hospitalization or prolongation of existing hospitalization • Persistent or significant disability or incapacity; • Congenital anomaly or birth defect • Other important medical event The investigator judged the relationship of an AE to study drug based on the timing of the AE relative to drug administration and whether or not other drugs, therapeutic interventions, or underlying conditions could provide an explanation for the event. The severity of an AE was evaluated by the investigator according to Common Terminology Criteria for Adverse Events (CTCAE Version 3.0), Japanese Clinical Oncology Group (JCOG) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death. Day 1 of study drug through 30 days after the last dose of study drug or discontinuation date; Up to data cut-off of 28 July 2015; maximum follow up time was 184.3 weeks
Secondary Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUC0-t) of Romidepsin in Phase 1 Area under the plasma concentration-time curve from time zero to the last quantifiable time point, calculated by the linear trapezoidal rule when concentrations are increasing and the logarithmic trapezoidal method when concentrations are decreasing. Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Area Under the Plasma Concentration-time Curve From Time Zero to Infinity (AUC8) of Romidepsin in Phase 1 Area under the plasma concentration-time curve from time zero extrapolated to infinity (AUC8) of romidepsin on Day 1; if possible the area under the concentration-time curve from time zero to infinity, calculated by the linear trapezoidal rule and extrapolated to infinity was calculated according to the following equation: AUC8 = AUCt + (Ct/ ?z ), where Ct is the last quantifiable concentration. Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Maximum Plasma Concentration (Cmax) of Romidepsin in Phase 1 The maximum observed plasma concentration of romidepsin (Cmax) obtained directly from the observed concentration versus time data Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Time to Maximum Plasma Concentration of Romidepsin (Tmax) in Phase 1 The time to first maximum observed plasma concentration of romidepsin after a single dose on Day 1. Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Terminal Phase Half-life of Romidepsin (t½) in Phase 1 The terminal phase half-life of romidepsin after a single dose on Day 1, calculated according to the following equation: t½ = 0.693/?z, where ?z is the terminal phase rate constant. Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration.
Secondary Apparent Total Clearance of Romidepsin (CL/F) of Romidepsin in Phase 1 The apparent total clearance of romidepsin after a single dose on Day 1, calculated as dose/AUC0-infinity. Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration.
Secondary Apparent Volume of Distribution (Vz/F) of Romidepsin in Phase 1 Apparent volume of distribution, was calculated according to the equation: Vd/F = (CL/F)/?z Day 1 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary AUC0-t, at Steady State (ss) of Romidepsin in Phase 1 at Cycle 1, Day 15 Area under the plasma concentration-time curve from time zero to the last quantifiable time point at steady state, calculated by the linear trapezoidal rule when concentrations are increasing and the logarithmic trapezoidal method when concentrations are decreasing Day 15 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Cmax, ss of Romidepsin in Phase 1 at Cycle 1, Day 15 Maximum observed concentration in plasma at steady state Day 15 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Tmax,ss of Romidepsin in Phase 1 at Cycle 1, Day 15 Observed time to first maximum plasma concentration at steady state Day 15 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Terminal Phase Half-life of Romidepsin (t½) in Phase 1 at Cycle 1, Day 15 The terminal phase half-life of romidepsin after a single dose on Day 15, calculated according to the following equation: t½ = 0.693/?z, where ?z is the terminal phase rate constant. Day 15 at Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration.
Secondary AUC0-t, Accumulation Ratio of Romidepsin in Phase 1, Cycle 1 Area under the plasma concentration-time curve from time zero to the last quantifiable time point; accumulation ratio calculated as AUC (0-t),ss/AUC (0-t) Day 1 and Day 15 in Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at the end of administration) hours after the start of administration, 0.25 (15 minutes), 0.5 (30 minutes), 1, 2, 4, 6, 20, and 44 hours after the end of administration
Secondary Cmax Accumulation Ratio of Romidepsin in Phase 1, Cycle 1 Cmax of Romidepsin: accumulation ratio based on Cmax calculated as Cmax,ss/Cmax Day 1 and Day 15 in Cycle 1; collected at 0 (pre-dose), 1, 2, 3, and 4 (at end of administration) hours after the start of administration, 0.25, 0.5, 1, 2, 4, 6, 20, and 44 hours after the end of administration. Day 8, Cycle 1, samples collected at 0 hour
Secondary The Percentage of Participants With Abnormal Q-wave and T Wave Intervals The time from the start of the Q-wave to the end of the T-wave QTc intervals greater than 450 msec post-baseline performed by centralized reviewer. The Bazett's (QTcB) and Fridericia (QTcF) correction were used as the standard clinical correction for calculating the heart rate-corrected QTc interval Median follow-up: 100 days; up to data cut-off of 28 July 2015
Secondary Percentage of PTCL Participants With the Best Response in Accordance With the Modified 2007 International Workshop Response Criteria as Assessed by IER Objective disease response in PTCL was defined as achieving a CR or PR based on the Modified 2007 IWC. A CR = a complete disappearance of all disease; lymph node mass regression to normal size on computerized tomography (CT) scan or negative on positron emission tomography (PET); non-palpable splenic and disappearance of liver nodules; infiltrate cleared on repeat bone marrow (BM), immunohistochemistry negative. PR = a reduction of measurable lesions; = 50% decrease in sum of the products of the greatest diameters (SPD) of up to 6 largest dominant masses, no enlargement in size of other nodes; = 50% decrease in SPD and no increase in liver or spleen. Tumor assessments performed every 2 months; median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Time to Response (TTR) for PTCL Participants With at Least a PR Based on the Modified 1999 IWC as Assessed by IER TTR for PTCL was defined as the time in days from first dose date to the first date of objective disease response. Median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Time to Response (TTR) for PTCL Participants With at Least a PR Based on the Modified 2007 IWC as Assessed by IER TTR for PTCL was defined as the time in days from first dose date to the first date of objective disease response. Median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Kaplan Meier Estimate of Duration of Response (DOR) for PTCL Responders Based on the Modified 1999 IWC as Assessed by the IER. DOR was defined as the number of days from the date of the first disease response (Complete, Unconfirmed Complete or Partial Response) until the date of progression, analyzed using Kaplan-Meier methods. Median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Kaplan Meier Estimate of Duration of Response (DOR) for PTCL Responders Based on the Modified 2007 IWC as Assessed by the IER. DOR was defined as the number of days from the date of the first disease response (Complete, Unconfirmed Complete or Partial Response) until the date of progression, analyzed using Kaplan-Meier methods. Median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Kaplan Meier Estimate of Time to Progression (TTP) in PTCL Participants Based on the 1999 IWC as Assessed by IER Time to progression (=50% increase from the nadir in the individual sum of the products of the diameters of any index lesion; the reappearance of pathology, enlargement of liver/spleen, or unequivocal progression of non-measurable disease or appearance of any new lesions) was defined as the duration from the date of the first study drug dose to the date of relapse or progression Median follow-up time was 100 days; up to the data cut-off of 28 July 2015
Secondary Kaplan Meier (K-M) Estimate of Time to Progression (TTP) in PTCL Participants Based on the Modified 2007 IWC as Assessed by IER Time to progression (=50% increase from the nadir in the individual sum of the products of the diameters of any index lesion; the reappearance of pathology, enlargement of liver/spleen, or unequivocal progression of non-measurable disease or appearance of any new lesions) was defined as the duration from the date of the first study drug dose to the date of relapse or progression Median follow-up time was 100 days; up to the data cut-off of 28 July 2015
See also
  Status Clinical Trial Phase
Active, not recruiting NCT03547700 - Study of Ixazomib and Romidepsin in Peripheral T-cell Lymphoma (PTCL) Phase 1/Phase 2
Completed NCT02788916 - A Retrospective Study of Clinical, Phenotypic and Genetic Factors of Peripheral T-Cell Lymphomas N/A
Completed NCT02567656 - Safety and Efficacy Study of a Dual PI3K Delta/Gamma Inhibitor in T-cell Lymphoma Phase 1
Completed NCT01142674 - T-Cell Project: Prospective Collection of Data in Patients With Peripheral T-Cell Lymphoma
Withdrawn NCT03355768 - Romidepsin Versus Combination of Romidepsin Plus Pralatrexate in PTCL Phase 3
Not yet recruiting NCT05006664 - Brentuximab Vedotin in Combination With CHEP in Patient With PTCL Phase 2
Terminated NCT00514722 - Pilot Study of Umbilical Cord Blood Transplantation in Adult Patient With Advanced Hematopoietic Malignancies N/A
Completed NCT01679860 - Intensive Chemo-immunotherapy as First Line Treatment in Adult Patients With Peripheral T- Cell Lymphoma Phase 2
Recruiting NCT06254495 - A Safety Study of SGN-35C in Adults With Advanced Cancers Phase 1
Completed NCT05137847 - A Study of Remitoro in Participants With Recurrent or Refractory Peripheral T Cell Lymphoma and Cutaneous T Cell Lymphoma (All Case Study)
Terminated NCT03601819 - Pacritinib in Relapsed/Refractory Lymphoproliferative Disorders Phase 1
Completed NCT03049449 - T Cells Expressing a Fully-Human Anti-CD30 Chimeric Antigen Receptor for Treating CD30-Expressing Lymphomas Phase 1
Completed NCT00211185 - A Study of ONTAK and CHOP in Newly Diagnosed, Peripheral T-Cell Lymphoma Phase 2
Not yet recruiting NCT03051581 - 18F-FDG PET/CT-based Prognostic Model for Predicting Outcome in Patients With Peripheral T-cell Lymphoma N/A
Not yet recruiting NCT03051568 - Evaluating 18F-FDG PET/CT With Liver SUVmax-based Criteria for Prognosis of Patients With Peripheral T-cell Lymphoma N/A
Completed NCT00646854 - Alemtuzumab and CHOP in T-cell Lymphoma Phase 3
Terminated NCT02535247 - Study of MK-3475 Alone or in Combination With Copanlisib in Relapsed or Refractory NK and T-cell Non-Hodgkin Lymphoma Phase 1/Phase 2
Recruiting NCT02364466 - Cohort of Peripheral T Cell Lymphoma N/A
Recruiting NCT02404571 - GDP in Frontline Chemotherapy for Patients With PTCL-NOS Phase 2
Completed NCT03742921 - ISTODAX® for Intravenous Infusion Drug Use Results Survey- Relapsed or Refractory Peripheral T-Cell Lymphoma