search
Back to results

PercutaneOus StEm Cell Injection Delivery Effects On Neomyogenesis in Dilated CardioMyopathy (The POSEIDON-DCM Study) (PoseidonDCM)

Primary Purpose

Non-ischemic Dilated Cardiomyopathy

Status
Completed
Phase
Phase 1
Locations
United States
Study Type
Interventional
Intervention
Autologous hMSCs
Allogeneic hMSCs
Sponsored by
Joshua M Hare
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Non-ischemic Dilated Cardiomyopathy focused on measuring Non ischemic dilated cardiomyopathy, Dilated cardiomyopathy, Heart failure, Cardiac Stem Cell Transplantation, Stem Cells

Eligibility Criteria

18 Years - 95 Years (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Major Inclusion Criteria:

  • Be ≥ 18 and < 95 years of age.
  • Provide written informed consent.
  • Diagnosis of nonischemic dilated cardiomyopathy.
  • Be a candidate for cardiac catheterization within 5 to 10 weeks of screening.
  • Been treated with appropriate maximal medical therapy for heart failure.
  • Ejection fraction below 40% and either a left ventricular end diastolic diameter (LVEDD) > 5.9cm in male subjects, an LVEDD of > 5.6cm in female subjects or left ventricular end diastolic volume index > 125 mL/m2
  • Be able to undergo an MRI or CT.

Major Exclusion Criteria:

  • Baseline glomerular filtration rate equal or < 45 ml/min/1.73m2.
  • Be eligible for or require standard-of-care surgical or percutaneous intervention for the treatment of nonischemic dilated cardiomyopathy.
  • Presence of a prosthetic aortic valve or heart constrictive device.
  • Presence of a prosthetic mitral valve.
  • Previous myocardial infarction (MI) as documented by a clinical history that will include an elevation of cardiac enzymes and/or ECG changes consistent with MI.
  • Diagnosis of nonischemic dilated cardiomyopathy due to valvular dysfunction, mitral regurgitation, tachycardia, or myocarditis.
  • Previous treatment for post-infarction left ventricular dysfunction including PCI and thrombolytic therapy.
  • Documented presence of a known LV thrombus, aortic dissection, or aortic aneurysm.
  • Documented presence of epicardial stenosis of 70% or greater in one or more major epicardial coronary arteries.
  • Documented presence of aortic stenosis (aortic stenosis graded as 1.5cm2 or less).
  • Documented presence of moderate to severe aortic insufficiency (echocardiographic assessment of aortic insufficiency graded as ≥+2).
  • Evidence of a life-threatening arrhythmia in the absence of a defibrillator (nonsustained ventricular tachycardia ≥ 20 consecutive beats or complete second or third degree heart block in the absence of a functioning pacemaker) or QTc interval > 550 ms on screening ECG.
  • AICD firing in the past 30 days prior to the procedure
  • Be eligible for or require coronary artery revascularization.
  • Diabetic with poorly controlled blood glucose levels and/or evidence of proliferative retinopathy.
  • Have a hematologic abnormality as evidenced by hematocrit < 25%, white blood cell < 2,500/ul or platelet values < 100,000/ul without another explanation.
  • Have liver dysfunction, as evidenced by enzymes (ALT and AST) greater than three times the ULN.
  • Have a coagulopathy condition = (INR > 1.3) not due to a reversible cause.
  • Known, serious radiographic contrast allergy.
  • Known allergies to penicillin or streptomycin.
  • Organ transplant recipient.
  • Have a history of organ or cell transplant rejection
  • Clinical history of malignancy within 5 years (i.e., patients with prior malignancy must be disease free for 5 years), except curatively-treated basal cell carcinoma, squamous cell carcinoma, or cervical carcinoma.
  • Non-cardiac condition that limits lifespan to < 1 year.
  • On chronic therapy with immunosuppressant medication.
  • Serum positive for HIV, hepatitis BsAg, or viremic hepatitis C.
  • Female patient who is pregnant, nursing, or of child-bearing potential and not using effective birth control.
  • Have a history of drug or alcohol abuse within the past 24 months.
  • Be currently participating (or participated within the previous 30 days) in an investigational therapeutic or device trial.

Sites / Locations

  • University of Miami School of Medicine

Arms of the Study

Arm 1

Arm 2

Arm Type

Active Comparator

Active Comparator

Arm Label

Autologous hMSCs

Allogeneic hMSCs

Arm Description

Group 1 (18 patients) Eighteen (18) patients will be treated with Auto-hMSCs: 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 x 108 (100 million) Auto-hMSCs.

Group 2 (18 patients) Eighteen (18) patients will be treated with allogeneic hMSCs (Allo-hMSCs): 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 x 108 (100 million) Auto-hMSCs.

Outcomes

Primary Outcome Measures

Incidence of Any Treatment-emergent Serious Adverse Events (TE-SAEs)
Incidence of TE-SAEs is defined as the composite of: death, non-fatal MI, stroke, hospitalization for worsening heart failure, cardiac perforation, pericardial tamponade, sustained ventricular arrhythmias (characterized by ventricular arrhythmias lasting longer than 15 seconds or with hemodynamic compromise), or any other potential late effects detected and corroborated by clinical presentation, laboratory investigations, image analysis and when necessary with biopsy from suspected target sites in the body.

Secondary Outcome Measures

Measurement of Changes in Peak VO2
Measurement of Peak oxygen consumption (Peak VO2) by treadmill determination during the 12 month follow-up period.
Measurement of Changes in 6 Minute Walk
Measurement of Six-minute walk test during the 12 month follow-up period
Measurement of Changes in Global Ejection Fraction
Measurement of regional left ventricular function, end diastolic and end systolic volume, measured by MRI, and or CT, and echocardiogram.
Measurement of Changes in New York Heart Association (NYHA)
Measurement of New York Heart Association (NYHA) functional class during the 12 month follow-up period.
Measurement of Changes in Minnesota Living With Heart Failure (MLHF) Questionnaire
Measurement of Minnesota Living with Heart Failure (MLHF) Questionnaire during the 12 month follow-up period. It measures the effects of symptoms, functional limitations, and psychological distress on an individual's quality of life. The response scale for all 21 items on the MLHF is based on a 6-point. The Maximum possible scores being 126 and the minimum 0. Higher scores indicate a worse or worsening quality of life, while lower scores or decreasing scores indicate a better quality of life.

Full Information

First Posted
June 29, 2011
Last Updated
January 19, 2018
Sponsor
Joshua M Hare
Collaborators
National Heart, Lung, and Blood Institute (NHLBI)
search

1. Study Identification

Unique Protocol Identification Number
NCT01392625
Brief Title
PercutaneOus StEm Cell Injection Delivery Effects On Neomyogenesis in Dilated CardioMyopathy (The POSEIDON-DCM Study)
Acronym
PoseidonDCM
Official Title
A Phase I/II, Randomized Pilot Study of the Comparative Safety and Efficacy of Transendocardial Injection of Autologous Mesenchymal Stem Cells Versus Allogeneic Mesenchymal Stem Cells in Patients With Non-ischemic Dilated Cardiomyopathy.
Study Type
Interventional

2. Study Status

Record Verification Date
January 2018
Overall Recruitment Status
Completed
Study Start Date
May 19, 2011 (Actual)
Primary Completion Date
August 28, 2016 (Actual)
Study Completion Date
August 28, 2017 (Actual)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor-Investigator
Name of the Sponsor
Joshua M Hare
Collaborators
National Heart, Lung, and Blood Institute (NHLBI)

4. Oversight

Studies a U.S. FDA-regulated Drug Product
Yes
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
The technique of transplanting progenitor cells into a region of damaged myocardium, termed cellular cardiomyoplasty1, is a potentially new therapeutic modality designed to replace or repair necrotic, scarred, or dysfunctional myocardium2-4. Ideally, graft cells should be readily available, easy to culture to ensure adequate quantities for transplantation, and able to survive in host myocardium; often a hostile environment of limited blood supply and immunorejection. Whether effective cellular regenerative strategies require that administered cells differentiate into adult cardiomyocytes and couple electromechanically with the surrounding myocardium is increasingly controversial and recent evidence suggests that this may not be required for effective cardiac repair. Most importantly, transplantation of graft cells should improve cardiac function and prevent adverse ventricular remodeling. To date, a number of candidate cells have been transplanted in experimental models, including fetal and neonatal cardiomyocytes5, embryonic stem cell-derived myocytes6, 7, tissue engineered contractile grafts8, skeletal myoblasts9, several cell types derived from adult bone marrow10-15, and cardiac precursors residing within the heart itself16. There has been substantial clinical development in the use of whole bone marrow and skeletal myoblast preparations in studies enrolling both post-infarction patients, and patients with chronic ischemic left ventricular dysfunction and heart failure. The effects of bone-marrow derived mesenchymal stem cells (MSCs) have also been studied clinically. Currently, bone marrow or bone marrow-derived cells represent highly promising modality for cardiac repair. The totality of evidence from trials investigating autologous whole bone marrow infusions into patients following myocardial infarction supports the safety of this approach. In terms of efficacy, increases in ejection fraction are reported in the majority of the trials. Non-ischemic dilated cardiomyopathy is a common and problematic condition; definitive therapy in the form of heart transplantation is available to only a tiny minority of eligible patients. Cellular cardiomyoplasty for chronic heart failure has been studied less than for acute MI, but represents a potentially important alternative for this disease.
Detailed Description
This is a Pilot Study, intended as a safety assessment prior to a full comparator study. In this Pilot Study, cells administered via the Biosense Webster MyoStar NOGA injection catheter system will be tested in 36 patients in two groups: Group 1 (18 patients) Eighteen (18) patients will be treated with Auto-hMSCs: 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 X 108 (100 million) Auto-hMSCs. Group 2 (18 patients) Eighteen (18) patients will be treated with Allo-hMSCs: 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 X 108 (100 million) Auto-hMSCs. The first three (3) patients in each group (Group 1 and Group 2) will not be treated less than 5 days apart and will each undergo full evaluation for 5 days to demonstrate there is no evidence of a procedure induced myocardial infarction or myocardial perforation prior to proceeding with the treatment of further patients. Patients will be randomized in a 1:1 ratio to one of the two groups. Treatment Strategies: Autologous hMSCs vs. Allogeneic hMSCs. The Study Team will record and maintain a detailed record of injection locations. If a patient is randomized to Groups 1 (Auto-hMSCs) and the Auto-hMSCs do not expand to the required dose of 1 X 108 cells, each injection will contain the maximum number of cells available. The injections will be administered transendocardially during cardiac catheterization using the Biosense Webster MyoStar NOGA Catheter System. For patients randomized to Group 1(Auto-hMSCs); the cells will be derived from a sample of the patient's bone marrow (obtained by iliac crest aspiration) approximately 4-6 weeks prior to cardiac catheterization. For patients randomized to Group 2 (Allo- hMSCs), the cells will be supplied from an allogeneic human mesenchymal stem cell source manufactured by the University of Miami. The Allo-hMSCs for patients in group 2 will be administered after all baseline assessments are completed with an expected range of 2 - 4 weeks post-randomization. Following cardiac catheterization and cell injections, patients will be hospitalized for a minimum of 2 days then followed at 2 weeks post-catheterization, and at month 2, 3, 6, and 12 to complete all safety and efficacy assessments.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Non-ischemic Dilated Cardiomyopathy
Keywords
Non ischemic dilated cardiomyopathy, Dilated cardiomyopathy, Heart failure, Cardiac Stem Cell Transplantation, Stem Cells

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1, Phase 2
Interventional Study Model
Parallel Assignment
Masking
Participant
Allocation
Randomized
Enrollment
37 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Autologous hMSCs
Arm Type
Active Comparator
Arm Description
Group 1 (18 patients) Eighteen (18) patients will be treated with Auto-hMSCs: 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 x 108 (100 million) Auto-hMSCs.
Arm Title
Allogeneic hMSCs
Arm Type
Active Comparator
Arm Description
Group 2 (18 patients) Eighteen (18) patients will be treated with allogeneic hMSCs (Allo-hMSCs): 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 x 108 (100 million) Auto-hMSCs.
Intervention Type
Biological
Intervention Name(s)
Autologous hMSCs
Other Intervention Name(s)
Biosense Webster MyoStar NOGA Injection Catheter System.
Intervention Description
Cells will be administered via the Biosense Webster MyoStar NOGA Injection Catheter System will be tested in 18 patients via transendocardial injection: Group 1 (18 patients) Eighteen (18) patients will be treated with Auto-hMSCs: 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 x 108 (100 million) Auto-hMSCs.
Intervention Type
Biological
Intervention Name(s)
Allogeneic hMSCs
Other Intervention Name(s)
Biosense Webster MyoStar NOGA Injection Catheter System.
Intervention Description
Cells will be administered via the Biosense Webster MyoStar NOGA Injection Catheter System will be tested in 18 patients via transendocardial injection: Group 2 (18 patients) Eighteen (18) patients will be treated with Allo-hMSCs: 20 million cells/ml delivered in a dose of 0.5 ml per injection x 10 injections for a total of 1 x 108 (100 million) Auto-hMSCs.
Primary Outcome Measure Information:
Title
Incidence of Any Treatment-emergent Serious Adverse Events (TE-SAEs)
Description
Incidence of TE-SAEs is defined as the composite of: death, non-fatal MI, stroke, hospitalization for worsening heart failure, cardiac perforation, pericardial tamponade, sustained ventricular arrhythmias (characterized by ventricular arrhythmias lasting longer than 15 seconds or with hemodynamic compromise), or any other potential late effects detected and corroborated by clinical presentation, laboratory investigations, image analysis and when necessary with biopsy from suspected target sites in the body.
Time Frame
One month post-catheterization
Secondary Outcome Measure Information:
Title
Measurement of Changes in Peak VO2
Description
Measurement of Peak oxygen consumption (Peak VO2) by treadmill determination during the 12 month follow-up period.
Time Frame
Baseline, 6 month and 12 month
Title
Measurement of Changes in 6 Minute Walk
Description
Measurement of Six-minute walk test during the 12 month follow-up period
Time Frame
Baseline, 6 month and 12 month
Title
Measurement of Changes in Global Ejection Fraction
Description
Measurement of regional left ventricular function, end diastolic and end systolic volume, measured by MRI, and or CT, and echocardiogram.
Time Frame
Baseline, 6 month and 12 month
Title
Measurement of Changes in New York Heart Association (NYHA)
Description
Measurement of New York Heart Association (NYHA) functional class during the 12 month follow-up period.
Time Frame
Baseline, 6 month and 12 month
Title
Measurement of Changes in Minnesota Living With Heart Failure (MLHF) Questionnaire
Description
Measurement of Minnesota Living with Heart Failure (MLHF) Questionnaire during the 12 month follow-up period. It measures the effects of symptoms, functional limitations, and psychological distress on an individual's quality of life. The response scale for all 21 items on the MLHF is based on a 6-point. The Maximum possible scores being 126 and the minimum 0. Higher scores indicate a worse or worsening quality of life, while lower scores or decreasing scores indicate a better quality of life.
Time Frame
Baseline, 6 month and 12 month

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Maximum Age & Unit of Time
95 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Major Inclusion Criteria: Be ≥ 18 and < 95 years of age. Provide written informed consent. Diagnosis of nonischemic dilated cardiomyopathy. Be a candidate for cardiac catheterization within 5 to 10 weeks of screening. Been treated with appropriate maximal medical therapy for heart failure. Ejection fraction below 40% and either a left ventricular end diastolic diameter (LVEDD) > 5.9cm in male subjects, an LVEDD of > 5.6cm in female subjects or left ventricular end diastolic volume index > 125 mL/m2 Be able to undergo an MRI or CT. Major Exclusion Criteria: Baseline glomerular filtration rate equal or < 45 ml/min/1.73m2. Be eligible for or require standard-of-care surgical or percutaneous intervention for the treatment of nonischemic dilated cardiomyopathy. Presence of a prosthetic aortic valve or heart constrictive device. Presence of a prosthetic mitral valve. Previous myocardial infarction (MI) as documented by a clinical history that will include an elevation of cardiac enzymes and/or ECG changes consistent with MI. Diagnosis of nonischemic dilated cardiomyopathy due to valvular dysfunction, mitral regurgitation, tachycardia, or myocarditis. Previous treatment for post-infarction left ventricular dysfunction including PCI and thrombolytic therapy. Documented presence of a known LV thrombus, aortic dissection, or aortic aneurysm. Documented presence of epicardial stenosis of 70% or greater in one or more major epicardial coronary arteries. Documented presence of aortic stenosis (aortic stenosis graded as 1.5cm2 or less). Documented presence of moderate to severe aortic insufficiency (echocardiographic assessment of aortic insufficiency graded as ≥+2). Evidence of a life-threatening arrhythmia in the absence of a defibrillator (nonsustained ventricular tachycardia ≥ 20 consecutive beats or complete second or third degree heart block in the absence of a functioning pacemaker) or QTc interval > 550 ms on screening ECG. AICD firing in the past 30 days prior to the procedure Be eligible for or require coronary artery revascularization. Diabetic with poorly controlled blood glucose levels and/or evidence of proliferative retinopathy. Have a hematologic abnormality as evidenced by hematocrit < 25%, white blood cell < 2,500/ul or platelet values < 100,000/ul without another explanation. Have liver dysfunction, as evidenced by enzymes (ALT and AST) greater than three times the ULN. Have a coagulopathy condition = (INR > 1.3) not due to a reversible cause. Known, serious radiographic contrast allergy. Known allergies to penicillin or streptomycin. Organ transplant recipient. Have a history of organ or cell transplant rejection Clinical history of malignancy within 5 years (i.e., patients with prior malignancy must be disease free for 5 years), except curatively-treated basal cell carcinoma, squamous cell carcinoma, or cervical carcinoma. Non-cardiac condition that limits lifespan to < 1 year. On chronic therapy with immunosuppressant medication. Serum positive for HIV, hepatitis BsAg, or viremic hepatitis C. Female patient who is pregnant, nursing, or of child-bearing potential and not using effective birth control. Have a history of drug or alcohol abuse within the past 24 months. Be currently participating (or participated within the previous 30 days) in an investigational therapeutic or device trial.
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Joshua M Hare, MD
Organizational Affiliation
University of Miami
Official's Role
Principal Investigator
Facility Information:
Facility Name
University of Miami School of Medicine
City
Miami
State/Province
Florida
ZIP/Postal Code
33136
Country
United States

12. IPD Sharing Statement

Plan to Share IPD
Undecided
Citations:
PubMed Identifier
21415390
Citation
Williams AR, Trachtenberg B, Velazquez DL, McNiece I, Altman P, Rouy D, Mendizabal AM, Pattany PM, Lopera GA, Fishman J, Zambrano JP, Heldman AW, Hare JM. Intramyocardial stem cell injection in patients with ischemic cardiomyopathy: functional recovery and reverse remodeling. Circ Res. 2011 Apr 1;108(7):792-6. doi: 10.1161/CIRCRESAHA.111.242610. Epub 2011 Mar 17.
Results Reference
background
PubMed Identifier
21392602
Citation
Trachtenberg B, Velazquez DL, Williams AR, McNiece I, Fishman J, Nguyen K, Rouy D, Altman P, Schwarz R, Mendizabal A, Oskouei B, Byrnes J, Soto V, Tracy M, Zambrano JP, Heldman AW, Hare JM. Rationale and design of the Transendocardial Injection of Autologous Human Cells (bone marrow or mesenchymal) in Chronic Ischemic Left Ventricular Dysfunction and Heart Failure Secondary to Myocardial Infarction (TAC-HFT) trial: A randomized, double-blind, placebo-controlled study of safety and efficacy. Am Heart J. 2011 Mar;161(3):487-93. doi: 10.1016/j.ahj.2010.11.024.
Results Reference
background
PubMed Identifier
27856208
Citation
Hare JM, DiFede DL, Rieger AC, Florea V, Landin AM, El-Khorazaty J, Khan A, Mushtaq M, Lowery MH, Byrnes JJ, Hendel RC, Cohen MG, Alfonso CE, Valasaki K, Pujol MV, Golpanian S, Ghersin E, Fishman JE, Pattany P, Gomes SA, Delgado C, Miki R, Abuzeid F, Vidro-Casiano M, Premer C, Medina A, Porras V, Hatzistergos KE, Anderson E, Mendizabal A, Mitrani R, Heldman AW. Randomized Comparison of Allogeneic Versus Autologous Mesenchymal Stem Cells for Nonischemic Dilated Cardiomyopathy: POSEIDON-DCM Trial. J Am Coll Cardiol. 2017 Feb 7;69(5):526-537. doi: 10.1016/j.jacc.2016.11.009. Epub 2016 Nov 14.
Results Reference
background
PubMed Identifier
31616309
Citation
Premer C, Wanschel A, Porras V, Balkan W, Legendre-Hyldig T, Saltzman RG, Dong C, Schulman IH, Hare JM. Mesenchymal Stem Cell Secretion of SDF-1alpha Modulates Endothelial Function in Dilated Cardiomyopathy. Front Physiol. 2019 Sep 24;10:1182. doi: 10.3389/fphys.2019.01182. eCollection 2019.
Results Reference
derived
PubMed Identifier
30005555
Citation
Tompkins BA, Rieger AC, Florea V, Banerjee MN, Natsumeda M, Nigh ED, Landin AM, Rodriguez GM, Hatzistergos KE, Schulman IH, Hare JM. Comparison of Mesenchymal Stem Cell Efficacy in Ischemic Versus Nonischemic Dilated Cardiomyopathy. J Am Heart Assoc. 2018 Jul 12;7(14):e008460. doi: 10.1161/JAHA.117.008460.
Results Reference
derived
PubMed Identifier
25354998
Citation
Mushtaq M, DiFede DL, Golpanian S, Khan A, Gomes SA, Mendizabal A, Heldman AW, Hare JM. Rationale and design of the Percutaneous Stem Cell Injection Delivery Effects on Neomyogenesis in Dilated Cardiomyopathy (the POSEIDON-DCM study): a phase I/II, randomized pilot study of the comparative safety and efficacy of transendocardial injection of autologous mesenchymal stem cell vs. allogeneic mesenchymal stem cells in patients with non-ischemic dilated cardiomyopathy. J Cardiovasc Transl Res. 2014 Dec;7(9):769-80. doi: 10.1007/s12265-014-9594-0. Epub 2014 Oct 30.
Results Reference
derived
Links:
URL
http://isci.med.miami.edu
Description
Interdisciplinary Stem Cell Institute University of Miami

Learn more about this trial

PercutaneOus StEm Cell Injection Delivery Effects On Neomyogenesis in Dilated CardioMyopathy (The POSEIDON-DCM Study)

We'll reach out to this number within 24 hrs