Anakinra in Preventing Severe Chimeric Antigen Receptor T-Cell Related Encephalopathy Syndrome in Patients With Recurrent or Refractory Large B-cell Lymphoma

June 15, 2023 updated by: Jonsson Comprehensive Cancer Center

IL-1 Receptor Antagonist to Prevent Severe Chimeric Antigen Receptor T-Cell Related Encephalopathy Syndrome

This phase II trial studies how well anakinra works in preventing severe chimeric antigen receptor T-cell-related encephalopathy syndrome after chimeric antigen receptor T-cell therapy in patients with large B-cell lymphoma that has come back or has not responded to treatment. Immunosuppressive therapy, such as anakinra, is used to decrease the body?s immune response, which may prevent severe chimeric antigen receptor T-cell-related encephalopathy syndrome.

Study Overview

Detailed Description

PRIMARY OBJECTIVES:

I. To determine if it is feasible to accrue a sufficient number of study participants at one site, in order to justify expanding the trial to three additional sites (pilot study).

II. To estimate the efficacy of anakinra in prevention of severe immune effector cell-associated neurotoxicity syndrome. syndrome (ICANS) (full study).

SECONDARY OBJECTIVES:

I. To estimate the impact that anakinra has on the efficacy of chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory lymphoma.

II. To estimate the rate of subsequent ICANS development in patients who receive anakinra for grade >= 3 cytokine release syndrome (CRS) in the absence of ICANS.

III. To estimate the duration of neurotoxicity in patients who receive anakinra.

IV. To estimate the duration of severe neurotoxicity in patients who receive anakinra.

V. To determine if anakinra causes persistent hepatotoxicity in patients receiving CAR T-cell for refractory lymphoma.

VI. to evaluate the overall toxicity of anakinra in patients receiving CAR T-cell therapy for refractory lymphoma.

EXPLORATORY OBJECTIVES:

I. To evaluate CRS and ICANS grade by using the American Society for Blood and Marrow Transplantation (ASBMT) 2018 consensus grading for adults.

II. To investigate changes in inflammatory markers including IL-1 and IL-6 in the peripheral blood during episodes of ICANS.

III. To describe the electroencephalogram (EEG) changes that characterize ICANS.

OUTLINE:

Patients receive standard lymphodepleting therapy including fludarabine and cyclophosphamide on days -5 to -3, then receive axicabtagene ciloleucel CAR T-cell infusion. Patients with clinical evidence of ICANS of any grade, or CRS >= grade 3 receive anakinra subcutaneously (SC) every 6-12 hours for 12-36 doses over 9 days in the absence of unacceptable toxicity.

After completion of study, patients are followed up at 30, 90, and 100 days, then at 6 months.

Study Type

Interventional

Enrollment (Estimated)

36

Phase

  • Phase 2

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Contact

Study Locations

    • California
      • Los Angeles, California, United States, 90095
        • Recruiting
        • UCLA / Jonsson Comprehensive Cancer Center
        • Contact:
        • Principal Investigator:
          • John M. Timmerman, M.D.

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Inclusion Criteria:

  • Patients with relapsed or refractory large B-cell lymphoma that has progressed on two prior lines of therapy, who meet the indication for the Food and Drug Administration (FDA)-approved therapy axicabtagene ciloleucel
  • Large B-cell lymphoma includes diffuse large B-cell lymphoma (DLBCL) not otherwise specified, primary mediastinal large B-cell lymphoma, high grade B-cell lymphoma, and DLBCL arising from follicular lymphoma
  • The above includes patients with progressive or stable disease as the best response to the most recent treatment regimen or disease progression within 12 months after autologous hematopoietic stem cell transplantation
  • Patients with central nervous system (CNS) involvement of large B-cell lymphoma that originated outside of the CNS will be included (not primary CNS lymphoma)
  • Serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) =< 2.5 upper limit of normal
  • Total bilirubin =< 2.0 mg/dL
  • Creatinine clearance > 30 mL/min based on Cockcroft-Gault formula
  • Patients with human immunodeficiency virus (HIV) who have an undetectable viral load will be included
  • Deemed competent to make medical decisions

Exclusion Criteria:

  • Patients who receive CAR T-cell therapy with a product other than axicabtagene ciloleucel
  • Primary CNS lymphoma
  • Transformed DLBCL from chronic lymphocytic leukemia (CLL)
  • Burkitt?s lymphoma
  • Bridging chemotherapy completed < 7 days prior to CAR T-cell lymphodepleting chemotherapy
  • In patients who receive bridging chemotherapy, positron emission tomography (PET)-computed tomography (CT) or CT of chest, abdomen, pelvis was not done after bridging chemotherapy prior lympho-depleting therapy
  • Most recent PET-CT or CT of all known disease is sites done more than 6 weeks prior to CAR T-cell infusion
  • Any individual CNS tumor mass > 2 cm
  • History of autologous hematopoietic stem cell transplantation administered less than 100 days prior to CAR T-cell infusion
  • History of allogeneic hematopoietic stem cell transplantation
  • Treatment with alemtuzumab within 6 months prior to leukapheresis, or treatment with clofarabine or cladribine within 3 months prior to leukapheresis
  • Less than 3 half-lives elapsed since receiving immune checkpoint inhibitor (pembrolizumab, ipilimumab, nivolumab, atezolizumab, etc.)
  • Presence of uncontrolled fungal, bacterial, or viral infection that require intravenous (IV) antimicrobial treatment
  • History of autoimmune disease resulting in end-organ damage, or autoimmune disease requiring systemic immunosuppressants or disease-modifying antirheumatic drug (DMARDs) within the past 6 months
  • Hypersensitivity to E. Coli-derived proteins
  • Patients with HIV who have a detectable viral load
  • Pregnant or nursing
  • Fertile women who decline use of contraception during the study period

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Prevention
  • Allocation: N/A
  • Interventional Model: Single Group Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Prevention (anakinra, CAR T-cell therapy)
Patients receive standard lymphodepleting therapy including fludarabine and cyclophosphamide on days -5 to -3, then receive axicabtagene ciloleucel CAR T-cell infusion. Patients with clinical evidence of ICANS of any grade, or CRS >= grade 3 receive anakinra SC every 6-12 hours for 12-36 doses over 9 days in the absence of unacceptable toxicity.
Given SC
Other Names:
  • Kinaret
  • Kineret
  • rIL-1ra
  • rIL1RN
Given via infusion
Other Names:
  • Fluradosa
Given via infusion
Other Names:
  • Yescarta
  • KTE C19
  • KTE-C19
  • KTE-C19 CAR
Given via infusion
Other Names:
  • Cytoxan
  • CTX
  • (-)-Cyclophosphamide
  • 2H-1,3,2-Oxazaphosphorine, 2-[bis(2-chloroethyl)amino]tetrahydro-, 2-oxide, monohydrate
  • Carloxan
  • Ciclofosfamida
  • Ciclofosfamide
  • Cicloxal
  • Clafen
  • Claphene
  • CP monohydrate
  • CYCLO-cell
  • Cycloblastin
  • Cycloblastine
  • Cyclophospham
  • Cyclophosphamid monohydrate
  • Cyclophosphamide Monohydrate
  • Cyclophosphamidum
  • Cyclophosphan
  • Cyclophosphane
  • Cyclophosphanum
  • Cyclostin
  • Cyclostine
  • Cytophosphan
  • Cytophosphane
  • Fosfaseron
  • Genoxal
  • Genuxal
  • Ledoxina
  • Mitoxan
  • Neosar
  • Revimmune
  • Syklofosfamid
  • WR- 138719
Given via infusion
Other Names:
  • 2-F-ara-AMP
  • Beneflur
  • Fludara
  • 9H-Purin-6-amine, 2-fluoro-9-(5-O-phosphono-.beta.-D-arabinofuranosyl)-
  • SH T 586

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Number of patients who met the eligibility criteria to receive and did receive anakinra
Time Frame: Up to 12 months
The study will be considered feasible if 8 study participants are enrolled over 12 months at University of California, Los Angeles.
Up to 12 months
Rate of severe chimeric antigen receptor T-cell-related encephalopathy syndrome (ICANS)
Time Frame: Up to 30 days
Will be defined as grade >= 3 neurotoxicity by Common Terminology Criteria for Adverse Events (CTCAE) version (v)4.03. The proportion of patients developing severe ICANS and the corresponding 90% binomial exact confidence interval (CI) will be reported.
Up to 30 days

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Objective response rate (ORR)
Time Frame: Up to 90 days
Will be defined as a complete response or partial response per the International Working Group criteria malignancy lymphoma based on positron emission tomography (PET)/computed tomography (CT) scan. To account for the adaptive nature of Simon?s two-stage design, final analysis of the objective response rate will report the uniformly minimum variance unbiased estimator and the corresponding p-value and 95% CI for the ORR.
Up to 90 days
Proportion of patients who received anakinra and develop ICAN
Time Frame: Up to 30 days
The proportion of patients who received anakinra in the absence of ICANS and then develop ICANS of any grade out of the total number of participants who received anakinra in the absence of ICANS will be evaluated. Will be summarized using descriptive statistics such as proportion, mean, standard deviation, median, as appropriate.
Up to 30 days
Duration of neurotoxicity
Time Frame: From first day of CRES of any grade to complete resolution of CRES, assessed up to 100 days
Defined as number of days that elapse from first day of ICANS of any grade to complete resolution of ICANS. Will be summarized using descriptive statistics such as proportion, mean, standard deviation, median, as appropriate.
From first day of CRES of any grade to complete resolution of CRES, assessed up to 100 days
Duration of neurotoxicity
Time Frame: From first day of ICANS of >= grade 3 to improvement of ICANS to < grade 3, assessed up to 100 days
Defined as number of days that elapse from first day of ICANS of >= grade 3 to improvement of ICANS to < grade 3 based on CTCAE v4.03. Will be summarized using descriptive statistics such as proportion, mean, standard deviation, median, as appropriate.
From first day of ICANS of >= grade 3 to improvement of ICANS to < grade 3, assessed up to 100 days
Persistent hepatotoxicity
Time Frame: Up to 100 days
Will be defined as grade > 2 aspartate aminotransferase (AST)/alanine aminotransferase (ALT) increased for at least 4 weeks duration according to CTCAE v4.03. Will be summarized using descriptive statistics such as proportion, mean, standard deviation, median, as appropriate.
Up to 100 days
Incidence of adverse events (AEs)
Time Frame: Up to 100 days
Will be based on CTCAE v4.03. Will be summarized using descriptive statistics such as proportion, mean, standard deviation, median, as appropriate. All AEs will be listed, documenting the course, outcome, severity, and relationship to the study treatment. Incidence rates of AEs and the proportion of subjects prematurely withdrawn from the study due to AEs will be shown.
Up to 100 days

Other Outcome Measures

Outcome Measure
Measure Description
Time Frame
ICANS grade
Time Frame: up to 30 days
Will evaluate ICANS grade by using the American Society for Blood and Marrow Transplantation 2018 consensus grading for adults.
up to 30 days
Cytokine release syndrome (CRS) grade
Time Frame: Up to 30 days
Will evaluate CRS grade by using the American Society for Blood and Marrow Transplantation 2018 consensus grading for adults.
Up to 30 days
Changes in inflammatory markers
Time Frame: Baseline up to 6 days following initiation of anakinra
Peak median serum blood and cerebral spinal fluid (CSF) levels of IL-1 and IL-6 will be summarized using descriptive statistics or contingency tables, as appropriate.
Baseline up to 6 days following initiation of anakinra
Changes in Electroencephalogram (EEG) that characterize ICANS: Slowing of EEG activity.
Time Frame: Baseline up to 100 days
Slowing of EEG activity including waveform, spectrum, spectrogram, and power in the slow (0.1-1 Hz), delta (1 to 4 Hz), theta (4 to 8 Hz) bands.
Baseline up to 100 days
Changes in Electroencephalogram (EEG) that characterize ICANS: focal slowing
Time Frame: Baseline up to 100 days
Changes in Electroencephalogram (EEG) that characterize ICANS: Focal regional slowing of the EEG activity within the frontal, temporal, parietal, or occipital region in the delta (<4Hz) and/ or theta (4-8 Hz) band.
Baseline up to 100 days
Changes in Electroencephalogram (EEG) that characterize ICANS: periodic discharge
Time Frame: Baseline up to 100 days
Generalized or lateralized or bilateral independent or multifocal periodic discharges (repetitive discharges with similar morphology and recurring at regular or near-regular inter-discharge intervals)
Baseline up to 100 days
Changes in Electroencephalogram (EEG) that characterize ICANS: rhythmic activity
Time Frame: Baseline up to 100 days
Generalized or lateralized or bilateral independent or multifocal Rhythmic delta activity (</= 4 Hz).
Baseline up to 100 days
Changes in Electroencephalogram (EEG) that characterize ICANS: Generalized or lateralized or bilateral independent or multifocal spike
Time Frame: Baseline up to 100 days
Generalized or lateralized or bilateral independent or multifocal spike (duration of the wave <70ms) or sharp (duration of the wave 70-200ms) discharges.
Baseline up to 100 days
Changes in Electroencephalogram (EEG) that characterize ICANS: seizures
Time Frame: Baseline up to 100 days
Electrographic seizure occurrence: Percentage of subjects who experience electrographic seizure while undergoing continuous-EEG monitoring.
Baseline up to 100 days

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Investigators

  • Principal Investigator: John M Timmerman, MD, UCLA / Jonsson Comprehensive Cancer Center

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

March 4, 2020

Primary Completion (Estimated)

June 1, 2024

Study Completion (Estimated)

June 1, 2025

Study Registration Dates

First Submitted

October 2, 2019

First Submitted That Met QC Criteria

December 17, 2019

First Posted (Actual)

December 20, 2019

Study Record Updates

Last Update Posted (Estimated)

June 16, 2023

Last Update Submitted That Met QC Criteria

June 15, 2023

Last Verified

February 1, 2023

More Information

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Progressive Disease

Clinical Trials on Anakinra

3
Subscribe