Stem Cell Injection in Cancer Survivors (SENECA)

October 13, 2020 updated by: Barry R Davis, The University of Texas Health Science Center, Houston

A Phase I, First-in-Human, Multicenter, Randomized, Double-Blinded, Placebo-Controlled Study of the Safety and Efficacy of Allogeneic Mesenchymal Stem Cells in Cancer Survivors With Anthracycline-Induced Cardiomyopathy

The primary purpose of this study is to examine the safety and feasibility of delivering allogeneic human mesenchymal stem cells (allo-MSCs) by transendocardial injection to cancer survivors with left ventricular (LV) dysfunction secondary to anthracycline-induced cardiomyopathy (AIC).

The secondary purpose of this study is to obtain preliminary evidence for therapeutic efficacy of allo-MSCs delivered by transendocardial injection to cancer survivors with LV dysfunction secondary to AIC.

Study Overview

Detailed Description

This phase I, randomized, placebo-controlled, trial will evaluate the safety and feasibility of allo-MSCs administered by transendocardial injection in thirty-seven subjects with anthracycline-induced cardiomyopathy (AIC). The first six subjects received allo-MSC therapy (open label) and were assessed for safety and feasibility of the study procedures. Following 1 month data review of each of the six subjects by the National Heart, Lung, and Blood Institute Gene and Cell Therapy Data Safety Monitoring Board; this was followed by a randomized, double-blind clinical trial enrolling thirty-one subjects. These subjects were randomized 1:1 to receive allo-MSCs or placebo. All subjects underwent cardiac catheterization and study product administration using the NOGA Myostar catheter injection system. Subjects are being followed at 1 day, 1 week, 1 month, 6 months, and 12 months post study product injection. All endpoints are assessed at the 6 and 12 month visits which will occur 180 ±30 days and 365 ±30 days, respectively, after the day of study product injection (Day 0). For the purpose of the safety evaluations and endpoint analysis, the Investigators will utilize an "intention-to-treat" study population. In addition, because this phase I study is the first cell therapy study in this population, at 12 months available standard-of-care medical records for cancer surveillance will be reviewed for cancer recurrence.

Study Type

Interventional

Enrollment (Actual)

46

Phase

  • Phase 1

Contacts and Locations

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

Study Locations

    • California
      • Stanford, California, United States, 94305
        • Stanford University School of Medicine
    • Florida
      • Gainesville, Florida, United States, 32610
        • University of Florida-Department of Medicine
      • Miami, Florida, United States, 33101
        • University of Miami-Interdiciplinary Stem Cell Institute
    • Indiana
      • Indianapolis, Indiana, United States, 46202
        • Indiana Center for Vascular Biology and Medicine
    • Kentucky
      • Louisville, Kentucky, United States, 40202
        • University of Louisville
    • Minnesota
      • Minneapolis, Minnesota, United States, 55407
        • Minneapolis Heart Institute Foundation
    • Texas
      • Houston, Texas, United States, 77030
        • Texas Heart Institute

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 to 79 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria

To participate, a subject MUST:

  1. Be ≥ 18 and < 80 years of age
  2. Be a cancer survivor with diagnosis of AIC
  3. Have an LVEF ≤ 45% by cMRI
  4. Be in NYHA class II-III
  5. Have received the initial diagnosis of AIC at least six months earlier and be on stable, optimally-tolerated therapy with beta-blockers, ACE inhibitors/ARBs, and/or aldosterone antagonists for 3 months, unless contraindicated
  6. Have a period of at least two years of clinical cancer-free state* and low likelihood of recurrence (a five-year risk of recurrence estimated at 30% or less), as determined by an oncologist, based on tumor type, response to therapy, and negative metastatic work-up at the time of diagnosis (*exceptions to this are carcinoma in situ or fully resected basal and squamous cell cancer of the skin.)
  7. Be a candidate for cardiac catheterization

Exclusion Criteria

To participate, a subject MUST NOT HAVE:

  1. A life expectancy <12 months
  2. A CT scan or baseline cardiac MRI showing new tumor or suspicious lymphadenopathy raising concern of malignancy
  3. Presence of obstructive CAD as determined via imaging within 5 years prior to study enrollment provided there have been no symptoms or evidence of CAD since the test
  4. Had a previous myocardial infarction
  5. A history of radiation therapy AND evidence of constrictive physiology and/or evidence of other patterns of non-ischemic cardiomyopathy on cardiac MRI (e.g., amyloidosis, sarcoidosis, hemochromatosis, pure radiation-induced cardiomyopathy, etc.) not consistent with AIC being the dominant etiology of heart failure
  6. Valvular heart disease including 1) mechanical or bioprosthetic heart valve; or 2) severe valvular (any valve) insufficiency/regurgitation within 12 months of consent.
  7. Aortic stenosis with valve area ≤ 1.5cm2
  8. A history of LV reduction surgery or cardiomyoplasty
  9. Evidence of cardiogenic shock
  10. A history of ischemic or hemorrhagic stroke within 90 days of baseline testing
  11. Liver dysfunction during baseline testing, as evidenced by enzymes (e.g., AST, ALT, alkaline phosphatase) greater than 3 times upper limit of normal
  12. Diabetes with poorly controlled blood glucose levels (HbA1c > 8.5%)
  13. An underlying autoimmune disorder or current immunosuppressive therapy (e.g., chronic corticosteroid, rheumatologic or immune modulating therapy) or likelihood of use of immunosuppressive therapy during participation in the trial (medications will be considered on a case by case basis)
  14. A baseline eGFR <35 ml/min/1.73m2
  15. A contrast allergy that cannot adequately be managed by premedication
  16. Received gene or cell-based therapy from any source within the previous 12 months
  17. A hematologic abnormality during baseline testing as evidenced by hemoglobin < 9 g/dl; hematocrit < 30%; absolute neutrophil count < 2,000 or total WBC count more than 2 times upper limit of normal; or platelet values < 100,000/ul
  18. Evidence of active systemic infection at time of study product delivery
  19. HIV and/or active HBV or HCV
  20. Coagulopathy (INR > 1.5) not due to a reversible cause (e.g., warfarin and/or Factor Xa inhibitors) (see Section 6.4 re: injection procedure and anticoagulation therapy) Note: Subjects who cannot be withdrawn from anticoagulation will be excluded.
  21. Presence of LV thrombus
  22. Presence of a pacemaker and/or ICD generator with any of the following limitations/conditions:

    • manufactured before the year 2000
    • leads implanted < 6 weeks prior to consent
    • non-transvenous epicardial or abandoned leads
    • subcutaneous ICDs
    • leadless pacemakers
    • any other condition that, in the judgment of device-trained staff, would deem an MRI contraindicated
  23. Pacemaker-dependence with an ICD (Note: pacemaker-dependent candidates without an ICD are not excluded)
  24. A cardiac resynchronization therapy (CRT) device implanted < 3 months prior to consent
  25. Other MRI contraindications (e.g. patient body habitus incompatible with MRI)
  26. An appropriate ICD firing or anti-tachycardia pacing (ATP) for ventricular fibrillation or ventricular tachycardia within 30 days of consent
  27. Ventricular tachycardia ≥ 20 consecutive beats without an ICD within 3 months of consent, or symptomatic Mobitz II or higher degree atrioventricular block without a functioning pacemaker within 3 months of consent
  28. A history of drug abuse (use of illegal "street" drugs except marijuana, or prescription medications not being used appropriately for a pre-existing medical condition) or alcohol abuse (≥ 5 drinks/day for ˃ 3 months), or documented medical, occupational, or legal problems arising from the use of alcohol or drugs within the past 24 months
  29. Cognitive or language barriers that prohibit obtaining informed consent or any study elements (interpreter permitted)
  30. Participation (currently or within the previous 30 days) in a cardiac related investigational therapeutic (including stem cell based therapies) or device trial
  31. Pregnancy, lactation, plans to become pregnant in the next 12 months, or is unwilling to use acceptable forms of birth control during study participation
  32. Any other condition that, in the judgment of the Investigator or Sponsor, would be a contraindication to enrollment, study product administration, or follow-up

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: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: Quadruple

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Allo-MSCs
Target dose of 100 million allo-MSCs
20 transendocardial injections of 0.4ml allo-MSCs administered to the left ventricle via NOGA Myostar injection catheter (single procedure)
Other Names:
  • Allogeneic Mesenchymal Stem Cells
Placebo Comparator: Placebo
Buminate solution
20 transendocardial injections of 0.4ml Buminate solution administered to the left ventricle via NOGA Myostar injection catheter (single procedure)
Other Names:
  • Buminate solution

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Proportion of Major Adverse Cardiac Events (MACE)
Time Frame: Baseline to 12 months
Proportion of adjudicated events including death, hospitalization for worsening heart failure, and/or other exacerbation of heart failure (non-hospitalization).
Baseline to 12 months
Proportion of Other Significant Clinical Events
Time Frame: Baseline to 12 months
Proportion of other significant adjudicated clinical events including: non-fatal stroke, non-fatal MI, coronary artery revascularization, ventricular tachycardia/fibrillation, pericardial tamponade, infectious myocarditis, hypersensitivity reaction, neoplasm, and/or other potential deleterious late effects.
Baseline to 12 months
Subjects With Events Precluding Their Receipt of Product
Time Frame: Randomization to SPI
Number and percent of subjects with events between randomization and study product injection (SPI) that preclude the subject from receiving product.
Randomization to SPI
Subjects Who Receive Less Than 20 Injections During SPI
Time Frame: During SPI procedure
Number and percent of subjects who receive less than 20 injections during SPI
During SPI procedure
Subjects Who Did Not Receive the Study Product (Either 100 Million Cells or Placebo)
Time Frame: During SPI procedure
Number and percent of subjects who did not receive the study product (either 100 million cells or placebo)
During SPI procedure
Subjects Who Have at Least One Cardiac MRI Endpoint Measure That is Uninterpretable
Time Frame: Baseline to 12 months
Number and percent of subjects who have at least one cardiac MRI endpoint measure that is uninterpretable due to issues related to the device, including, but not limited to, inability to undergo the procedure.
Baseline to 12 months
Subjects Who Fail to Complete Follow-up
Time Frame: Baseline to 12 months
Number and percent of subjects who fail to complete follow up
Baseline to 12 months

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Change From Baseline in Left Ventricular Ejection Fraction (LVEF)
Time Frame: Baseline to 12 months
Change in left ventricular ejection fraction as assessed via cardiac MRI.
Baseline to 12 months
Change From Baseline in Left Ventricular Ejection Fraction (LVEF)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Global Strain (HARP MRI)
Time Frame: Baseline to 12 months
Change in global circumferential strain as assessed via cardiac MRI
Baseline to 12 months
Change From Baseline in Global Strain (HARP MRI)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Regional Strain (HARP MRI)
Time Frame: Baseline to 12 months
Change in regional longitudinal strain as assessed via cardiac MRI
Baseline to 12 months
Change From Baseline in Regional Strain (HARP MRI)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Left Ventricular End Diastolic Volume Index (LVEDVI)
Time Frame: Baseline to 12 months
Change in left ventricular end diastolic volume index as measured via cardiac MRI
Baseline to 12 months
Change From Baseline in Left Ventricular End Diastolic Volume Index (LVEDVI)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Left Ventricular End Systolic Volume Index (LVESVI)
Time Frame: Baseline to 12 months
Change in left ventricular end systolic volume index as assessed via cardiac MRI
Baseline to 12 months
Change From Baseline in Left Ventricular End Systolic Volume Index (LVESVI)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Left Ventricular Sphericity Index
Time Frame: Baseline to 12 months
Change in Left Ventricular Sphericity Index as assessed by cardiac MRI. Sphericity index is the ratio of the long and short axis measurements of the left ventricle.
Baseline to 12 months
Change From Baseline in Left Ventricular Sphericity Index-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)

The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.

Sphericity index is the ratio of the long and short axis measurements of the left ventricle.

Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Area of Injury
Time Frame: Baseline to 12 months
Change in the scar percent (scar mass normalized to left ventricular mass) as assessed via cardiac MRI.
Baseline to 12 months
Change From Baseline in Area of Injury-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Exercise Tolerance (Six Minute Walk Test)
Time Frame: Baseline to 12 months
Change in the distance walked (in meters) as measured by the six minute walk test. Two walk tests were completed at each endpoint visit (separated by 30 min). The average distance of the two walk tests will be used for analysis.
Baseline to 12 months
Change From Baseline in Exercise Tolerance (Six Minute Walk Test)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
Change in the distance walked (in feet) as measured by the six minute walk test. Two walk tests were completed at each endpoint visit (separated by 30 min). The average distance of the two walk tests will be used for analysis. The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in Minnesota Living With Heart Failure Questionnaire Score
Time Frame: Baseline to 12 months
Change in the quality of life summary score as measured by the Minnesota Living with Heart Failure Questionnaire. Minimum and maximum scores for scale are 0 and 105 respectively. Lower scores indicative of better outcome.
Baseline to 12 months
Change From Baseline in Minnesota Living With Heart Failure Questionnaire Score-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)

The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.

Minimum and maximum scores for scale are 0 and 105 respectively. Lower scores indicative of better outcome.

Assessed as a trajectory (baseline, 6 months, and 12 months)
Change From Baseline in N-Terminal Pro-Brain Natriuretic Peptide (NT-proBNP)
Time Frame: Baseline to 12 months
Change in N-Terminal pro-Brain Natriuretic Peptide (NT-proBNP) as measured via laboratory blood draw
Baseline to 12 months
Change From Baseline in N-Terminal Pro-Brain Natriuretic Peptide (NT-proBNP)-Trajectory
Time Frame: Assessed as a trajectory (baseline, 6 months, and 12 months)
The change in this measure over time is assessed using a repeated measures linear regression model of trajectory (change over time). If there is no interaction between change over time and treatment a single calculated value is the slope (the change per six months) from the model. If there is an interaction, the p-value for interaction is presented along with two calculated values representing the slope (the change per six months) for each treatment arm from the model.
Assessed as a trajectory (baseline, 6 months, and 12 months)
Cumulative Days Alive and Out of Hospital for Heart Failure
Time Frame: Baseline to End of 12 Month Visit Window (i.e. 395 days after intervention)
Days alive and out of hospital for heart failure during the study evaluation period. Subjects were allotted a visit window extending 30 days past their anticipated 12-month visit (i.e., 395 days).
Baseline to End of 12 Month Visit Window (i.e. 395 days after intervention)

Collaborators and Investigators

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

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

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)

August 1, 2016

Primary Completion (Actual)

November 1, 2019

Study Completion (Actual)

April 20, 2020

Study Registration Dates

First Submitted

July 23, 2015

First Submitted That Met QC Criteria

July 23, 2015

First Posted (Estimate)

July 27, 2015

Study Record Updates

Last Update Posted (Actual)

November 5, 2020

Last Update Submitted That Met QC Criteria

October 13, 2020

Last Verified

October 1, 2020

More Information

Terms related to this study

Other Study ID Numbers

  • HSC-SPH-15-0443
  • 5UM1HL087318 (U.S. NIH Grant/Contract)

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 Cardiomyopathy Due to Anthracyclines

Clinical Trials on Allo-MSCs

3
Subscribe