The CardiAMP Heart Failure trial: A randomized controlled pivotal trial of high-dose autologous bone marrow mononuclear cells using the CardiAMP cell therapy system in patients with post–myocardial infarction heart failure: Trial rationale and study design

The CardiAMP Heart Failure trial: A randomized controlled pivotal trial of high-dose autologous bone marrow mononuclear cells using the CardiAMP cell therapy system in patients with post–myocardial infarction heart failure: Trial rationale and study design

Accepted Manuscript The CardiAMP Heart Failure trial: A Randomized Controlled Pivotal Trial of High Dose Autologous Bone Marrow Mononuclear Cells Usin...

954KB Sizes 1 Downloads 24 Views

Accepted Manuscript The CardiAMP Heart Failure trial: A Randomized Controlled Pivotal Trial of High Dose Autologous Bone Marrow Mononuclear Cells Using the CardiAMP Cell Therapy System in Patients with Post Myocardial Infarction Heart Failure: Trial Rationale and Study Design

Amish N. Raval MD FSCAI FACC, Thomas D. Cook, Henricus J. Duckers, Peter V. Johnston, Jay H. Traverse, William T. Abraham, Peter A. Altman, Carl J. Pepine PII: DOI: Reference:

S0002-8703(18)30100-5 doi:10.1016/j.ahj.2018.03.016 YMHJ 5655

To appear in: Received date: Accepted date:

13 December 2017 24 March 2018

Please cite this article as: Amish N. Raval MD FSCAI FACC, Thomas D. Cook, Henricus J. Duckers, Peter V. Johnston, Jay H. Traverse, William T. Abraham, Peter A. Altman, Carl J. Pepine , The CardiAMP Heart Failure trial: A Randomized Controlled Pivotal Trial of High Dose Autologous Bone Marrow Mononuclear Cells Using the CardiAMP Cell Therapy System in Patients with Post Myocardial Infarction Heart Failure: Trial Rationale and Study Design. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Ymhj(2018), doi:10.1016/j.ahj.2018.03.016

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

The CardiAMP Heart Failure trial: A Randomized Controlled Pivotal Trial of High Dose Autologous Bone Marrow Mononuclear Cells Using the CardiAMP Cell Therapy System in Patients with Post Myocardial Infarction Heart Failure: Trial Rationale and Study

T

Design

CR

IP

RCT# NCT02438306

US

Amish N. Ravala, Thomas D. Cookb, Henricus J. Duckersc, Peter V. Johnstond, Jay H. Traversee, William T. Abrahamf, Peter A. Altmanc, Carl J. Pepineg a

Division of Cardiovascular Medicine, Department of Medicine and Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison

c

Biocardia Inc. San Carlos, CA.

AN

b

d

Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, USA

ED

e

M

Division of Cardiovascular Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD

f

PT

Departments of Medicine, Physiology, and Cell Biology, Division of Cardiovascular Medicine, and the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio. g

Division of Cardiovascular Medicine, Department of Medicine, University of Florida,Gainesville, FL

CE

Short Title: Raval. The CardiAMP Heart Failure Trial Rationale and Study Design

AC

Corresponding Author:

Amish N. Raval MD, FSCAI, FACC, FAHA Associate Professor of Medicine Division of Cardiovascular Medicine Department of Medicine and Biomedical Engineering University of Wisconsin School of Medicine and Public Health H4/568 Clinical Sciences Center 600 Highland Ave, Madison, WI 53792-3248, USA Office:(608) 263-0836 Cell: (608) 347-8074 Fax: (608) 263-0405 Email: [email protected]

ACCEPTED MANUSCRIPT

Journal Subject Terms: Cell Therapy, Stem cells, Heart Failure, Chronic Ischemic Heart Disease, Treatment. Abstract Background: Heart failure following myocardial infarction is a common, disabling and deadly

IP

T

condition. Direct injection of autologous bone marrow mononuclear cells (BM MNCs) into the myocardium may result in improved functional recovery, relieve symptoms and improve other

CR

cardiovascular outcomes.

US

Methods: CardiAMP-HF is a randomized, double-blind, sham-controlled, pivotal trial designed to investigate the safety and efficacy of autologous BM MNC treatment for patients

AN

with medically refractory and symptomatic ischemic cardiomyopathy. The primary endpoint is

M

change in six-minute walk distance adjusted for major adverse cardiovascular events at 12 months following treatment. Particularly novel aspects of this trial include a cell potency assay

ED

to screen subjects who have bone marrow cell characteristics that suggest a favorable response to treatment, a point of care treatment method, a high target dose of 200 million

CE

high cell retention.

PT

cells and an efficient transcatheter intramyocardial delivery method that is associated with

Conclusions: This novel approach may lead to a new treatment for those with ischemic

AC

heart disease suffering from medically refractory heart failure. Clinical Trial Registration: https://clinicaltrials.gov/ct2/show/NCT02438306 Key words: Cell, Heart Failure, Treatment, Trial, Catheter

2

ACCEPTED MANUSCRIPT

Introduction Heart failure following myocardial infarction affects ~5 million Americans and is a common reason for hospitalization, which impairs quality of life and consumes enormous health care

T

resources1. The mortality rate of these patients is high and similar to that of advanced cancer.

IP

Guideline-based therapy, using medications that block maladaptive neurohormonal pathways,

CR

is often only partially effective and not universally tolerated. Left ventricular assist devices and heart transplant may be offered in advanced heart failure, but device failure, stroke, infection,

US

and organ shortages limit these approaches. Cell based therapy represents an innovative approach to recover contractile heart function and improve clinical outcomes. However,

AN

despite two decades of investigation, no Food and Drug Administration (FDA) cell treatments

M

for cardiovascular disease have been approved.

Adult bone marrow contains a large reservoir of stem and progenitor cells capable of

ED

differentiating into hematopoietic, endothelial and mesenchymal lineages. In addition, numerous pre-clinical cardiac studies have shown that paracrine signaling by bone marrow-

PT

derived stem cells promotes microcirculatory adaptation, immune modulation and

CE

cytoprotection through anti-apoptotic effects, facilitating post-infarct cardiac recovery, in part via recruitment of endogenous cardiac stem/progenitor cells2-12. Improvements in left

AC

ventricular myocardial perfusion and contractile function have been demonstrated in small and large animal models following intramyocardial injection of bone marrow-derived stem cells13-19. These encouraging pre-clinical findings paved the way for several Phase I and II clinical trials of autologous bone marrow cell therapy for chronic ischemic heart disease and postmyocardial infarct recovery20-35. For example, the Transendocardial Autologous Bone Marrow in Myocardial Infarction (TABMMI) heart failure trial treated 20 chronic ischemic heart failure patients with autologous bone marrow derived mononuclear cells (BM MNC) using a 3

ACCEPTED MANUSCRIPT

transendocardial helical injection catheter24, 35. Left ventricular ejection fraction and exercise tolerance were improved at up to 5 years follow-up, with no treatment emergent major adverse events. Subsequently, the Transendocardial Autologous Cells in Ischemic Heart Failure Trial (TAC-HFT) compared autologous BM MNC treatment and autologous bone

T

marrow derived mesenchymal stem cell treatment with two corresponding placebo arms in

IP

patients with ischemic heart failure32. Transendocardial delivery of BM MNC was shown to be

CR

safe at 30 days and 1 year. When the BM MNC treated group (n=19) was compared to a pooled placebo group (n=20) in the study, quality of life as measured by the Minnesota Living

US

with Heart Failure Questionnaire (MLWHFQ) and six minute walk distance (6MWD) were

AN

improved at 12 months, although the latter was not statistically significant (p = 0.005 and p = 0.11, respectively). In a separate analysis34 in which the BM MNC treated group (n=19) was

M

compared to the matched placebo group (n=10), there was significant improvement in 6MWD

ED

at 12 months [BM MNC group (+14.3 meters, CI -15.4 to 43.9) versus the placebo group (42.0 meters, CI -102.1 to 18.1), P=0.049]. In this analysis, the MLWHFQ score also improved

PT

in the BM MNC group compared to a deterioration in the corresponding placebo group at 12 months [BM MNC group -7.7, SD 24.8 versus Placebo group +9.7, SD 24.8 (P=0.038)].

CE

Likewise, all other pre-specified endpoints favored the BM MNC group whereas the placebo

AC

treated patients deteriorated over 12 month follow-up. Finally, multiple imputation and rank adjustment for missing 6MWD results due to intervening major adverse cardiovascular events (MACE) was applied to the data, revealing greater statistical confidence of the between group differences in 6MWD (refer to SUPPLEMENT 1 for more detail). The safety of the treatment approach combined with the improvements in 6MWD and quality of life observed in the Phase I (TABMMI) and Phase II (TAC-HFT) trials provide compelling evidence to support further study of BM MNC treatment of patients with chronic heart failure due to ischemic heart disease (IHD). 4

ACCEPTED MANUSCRIPT

A 2016 Cochrane meta-analysis of 38 randomized-controlled trials assessing effects of BM MNCs in 1907 heart failure patients with IHD found that BM MNC treatment significantly reduced all-cause mortality, non-fatal myocardial infarction, and arrhythmias at 12 month follow-up, although heterogeneity among the trials limited the robustness of the analysis36, 37.

T

The totality of clinical trial evidence to date supports that intramyocardial delivery of

CR

controlled trials are required to establish clinical efficacy.

IP

autologous bone marrow mononuclear cells is safe; however, more robust randomized-

US

The pivotal CardiAMP-HF trial is a novel approach to the problem of post myocardial infarction heart failure. It will assess patients with New York Heart Association (NYHA) functional class

AN

II-III heart failure, who are selected based on their bone marrow potency, whether intramyocardial injection of high dose autologous bone marrow mononuclear cells will improve

M

ambulatory function and quality of life, and prevent MACE. Prior to entry, patients will be

ED

screened using a cell potency assay that measures the stem cell characteristics of their bone marrow. Those subjects meeting the selection criteria will have their BM MNCs harvested and

PT

immediately isolated using a point-of-care device in the cardiac catheterization laboratory.

CE

The CardiAMP-HF cell product will be then be delivered into the myocardium using a novel

Methods:

AC

and efficient helical needle tipped transendocardial delivery catheter.

CardiAMP-HF is a pivotal, randomized controlled trial to investigate the safety and efficacy of the CardiAMP cell therapy system (BioCardia, San Carlos, CA) in bone marrow potency selected patients with post-myocardial infarction NYHA functional class II-III heart failure (NCT02438306) enrolled in up to 40 clinical centers in the United States. The trial has two cohorts: 1) a roll-in phase with a prospective, open-label, uncontrolled, patient cohort to evaluate on-site physician and coordinator training in the various procedures/tests and ensure 5

ACCEPTED MANUSCRIPT

timely transfer of materials and information between sites and core laboratories to create best practices before opening additional enrolling sites (n=10), and 2) the prospective, doubleblinded, randomized (3:2 treatment vs. sham) cohort to establish clinical efficacy (n=250) (Figure 1). The trial is being conducted under FDA Investigational Device Exemption (IDE)

T

number 16057. Enrollment is anticipated to be completed by June 2019, and study

CR

IP

completion by June 2021. Study Population

US

Subjects between 21 and 90 years of age, with chronic, symptomatic ischemic left ventricular dysfunction and NYHA functional class II-III are eligible for enrollment. Written informed

AN

consent is obtained by members of the investigational site with institutional review board (IRB)

M

approval. Chronic ischemic left ventricular dysfunction is defined as a left ventricular ejection fraction (EF) of 20-40% after a remote (>6 months) myocardial infarction (MI). All subjects are

ED

required to be receiving an “optimized” regimen of guideline-directed-medical therapy for at least 3 months prior to treatment. All subjects must be ambulatory and capable of performing

PT

a 6MWD test with a distance >100 meters but < 450 meters. Key inclusion and exclusion

CE

criteria for the CardiAMP HF trial are summarized in Table 1.

AC

Cell Potency Assay

All subjects enrolled in the trial first undergo a 5mL bone marrow aspirate from the iliac crest during the screening period, which is 4 to 45 days prior to randomization. The aspirate is shipped overnight to an independent Cell Analysis Core Laboratory (Center for Cell and Gene Therapy, Baylor University, Houston TX). The cellular aspirate is tested for pre-defined markers that have pro-angiogenic and cardio-reparative potential. The results are expressed as a Cell Potency Assay (CPA) score. All investigators are masked to the details and specific

6

ACCEPTED MANUSCRIPT

results of this assay in any given patient, other than whether or not the patient’s bone marrow qualifies. This proprietary CPA is detailed in the approved FDA IDE. The CardiAMP HF bone marrow CPA is derived from observations in prior ischemic heart

T

disease cell therapy trials. For example, a lower limit of the effective dose threshold for CD34+

IP

cells is one component of the assay. CD34+ cells are associated with favorable angiogenic and regenerative properties in a number of pre-clinical and clinical cardiovascular disease

CR

trials27, 28, 38-41. In the FOCUS trial31, which tested intramyocardial injections of unselected BM

US

MNCs in ischemic heart failure patients, it was observed that those patients who received higher doses of CD34+ cells were correlated with further improvements in left ventricular

AN

ejection fraction (LVEF). Every 3% higher level of CD34+ cells was associated with, on average, a 3.0% greater absolute unit increase in LVEF. Furthermore, transendocardial

M

administration of CD34+ cells were associated with improvements in exercise tolerance in the

ED

ACT34+ chronic myocardial ischemia trial28, the RENEW trial42, and in recently published patient level pooled analysis of a combination of chronic myocardial ischemia trials43. Based

PT

on calculations performed on these prior data, it is estimated that ~70% of subjects will meet

CE

the pre-specified CPA criteria, and therefore, qualify for enrollment. Enrolled subjects will undergo a second, larger volume, bone marrow aspirate to obtain the treatment product as

AC

described below in Treatment Procedure. Subjects who fail to meet the CPA criteria will not be enrolled in the randomized phase of the trial. Treatment Procedure All subjects who meet screening criteria will undergo aspiration of 60 mL of bone marrow from the iliac crest under local anesthesia and mild sedation if required. The bone marrow aspirate is immediately processed using a point-of-care system (CardiAMP™ Cell Separator, Biocardia, San Carlos, CA) in the cardiac catheterization laboratory (Figure 2). The process is a density7

ACCEPTED MANUSCRIPT

tuned dual buoy separation to isolate the BM MNC fraction that typically takes ~25 minutes. Patients will receive a dose targeted to yield ~200 million autologous bone marrow mononuclear cells, identical to the phase II TAC-HFT trial32.

T

Immediately following bone marrow aspiration, patients are randomized 3:2 to either the BM

IP

MNC therapy group or a sham procedure group. Randomization will be stratified for patients

CR

who have cardiac resynchronization therapy (CRT) at entry using a computerized block randomization method.

US

All subjects undergo percutaneous femoral artery access, left heart catheterization and ventriculography in two orthogonal projections. Subjects randomized to active treatment then

AN

undergo 10 transendocardial injections of 0.5 ml high dose BM MNC in the peri-infarct

M

myocardial segments. Target segments are selected based on a 17 segment bulls’ eye maps depicting myocardial wall thickness and local wall motion, determined by baseline contrast–

ED

enhanced echocardiography. Injections are performed using the Helix™ dual lumen,

PT

intramyocardial injection catheter and the Morph® deflectable guide catheter (BioCardia, San Carlos, CA; Figure 3). Diluted contrast solution is infused through a separate contrast lumen in

CE

the Helix catheter to verify engagement of the target myocardium. Subjects randomized to sham treatment will likewise undergo bone marrow aspiration, placement of an arterial sheath,

AC

and left heart catheterization with ventriculography. Thereafter, the interventionalist and staff will follow a scripted sham protocol to mimic the activities of an injection procedure to maintain subject blinding. After the treatment, a separate team of physicians and study personnel who are blinded to the treatment assignment will perform the follow-up visits with the subject. Outcomes The primary outcome is a composite that combines change in 6MWD with missing values ranked according to the severity of heart failure defined as first occurrence of a major 8

ACCEPTED MANUSCRIPT

cardiovascular event (MACE) among all-cause mortality, hospitalization for worsening heart failure, non-fatal recurrent myocardial infarction, placement of a left ventricular assist device, or heart transplantation. The 6MWD will be measured at baseline, and at intervals of 3 months over the first 12 months, and then again at 24 months. For each time point, two 6MWD

T

measurements will be performed under identical conditions, with at least a 1-hour recovery

IP

between measurements. The longest 6MWD distance at each time point will be used for the

CR

analysis. If the difference between the two 6MWD tests is >10%, a third test will be performed

US

and the longest 6MWD will be used for analysis.

Secondary outcomes will be adjudicated following a hierarchical scheme and gatekeeper

AN

approach. These include: i) overall 12-month survival (non-inferiority), ii) freedom from heart failure MACE at 12-months; non-inferiority), iii) change in quality of life as measured by the

M

Minnesota Living with Heart Failure Questionnaire44 at 12-months (superiority), iv) Time to first

ED

MACE at 12-months (superiority), and v) overall 12 month survival (superiority). Additional secondary outcomes will include treatment-emergent serious adverse events at 30-

PT

days, days alive out of hospital, New York Heart Association (NYHA) heart failure Functional

CE

Class, echocardiographic measures of change in ejection fraction and left ventricular endsystolic and end-diastolic dimensions. Echocardiographic analysis is performed by a blinded

AC

independent imaging core laboratory (Yale Cardiovascular Research Group, New Haven, CT). All clinical events are adjudicated by an independent clinical events committee (CEC). An independent data safety monitoring committee monitors the study for safety outcomes and futility in achieving the primary efficacy outcomes. Importantly, prior transendocardial injection studies have shown modestly elevated cardiac biomarkers up to 48-hours after the procedure, without any clinical evidence of an acute coronary syndrome, including signs or symptoms, electrocardiographic changes or novel wall motion abnormalities on echocardiography. 9

ACCEPTED MANUSCRIPT

Therefore, the clinical relevance of these modestly elevated biomarkers following transendocardial injection is unclear45. The independent CEC has chosen to define “peri procedural infarction” as an adverse event occurring within the first 48-hours after the study procedure, in association with at least a 5 x the upper limit of normal (ULN) increase in CK-MB

T

and/or 35 x ULN increase in Troponin T, in combination with evidence of prolonged ischemia,

IP

as demonstrated with prolonged chest pain, electrocardiographic (ST changes, new Q

CR

waves), or imaging evidence of new regional wall motion abnormalities according to the Joint ESC/ACCF/AHA/WHF Taskforce for the Universal Definition of Myocardial Infarction46. All

US

peri-procedural biomarker changes will be captured, but only those that meet these criteria will

AN

be considered adverse events and will be evaluated on a case-to-case basis by the CEC.

M

Statistical Analysis Plan

The primary endpoint is a comparison of a composite of the change in distance walked in 6-

ED

minutes and heart failure MACE between the treatment group and the sham-control group at 12 months (superiority outcome). If the primary endpoint is significantly improved (p<0.05),

PT

then a series of pre-specified secondary endpoints will be analyzed in a hierarchical fashion

CE

using a gatekeeper approach (described in the “Statistical Analysis Plan” in SUPPLEMENT 2). These secondary endpoints are ranked as follows: 1) overall survival at 12-months as a non-

AC

inferiority outcome, 2) freedom from MACE defined as the composite of all-cause death, hospitalization for worsening heart failure, non-fatal recurrent myocardial infarction, placement of a left ventricular assist device or heart transplant at 12-months as a non-inferiority outcome, 3) change in quality of life as measured by Minnesota Living with Heart Failure Questionnaire at 12 months as a superiority outcome, 4) time to first MACE at 12 months as a superiority outcome and 5) overall survival at 12 months as a superiority outcome. Hierarchical

10

ACCEPTED MANUSCRIPT

secondary endpoints will be evaluated sequentially, conditional on all treatment comparisons for previous endpoints reaching statistical significance. It is expected that a proportion of subjects will either die or experience a non-lethal MACE

T

event that will preclude the assessment of the 6MWD. Therefore, the statistical analysis plan

IP

is designed to account for these missing variables. The MACE component of the primary

CR

endpoint will be ranked according to the severity of clinical outcome, with the inability to perform the 6MWD due to death of the subject awarded as lowest rank. Progressive ranks are

US

assigned for intervening non-lethal MACE events that prevent the performance of the 6MWD test. The highest rank would be for those individuals who achieve a successful 6MWD without

AN

intervening death or MACE. The analysis will use a semi-parametric proportional odds model with terms for treatment and baseline 6MWD. This approach has greater power than using

M

change in 6MWD and the adjustment for baseline implicitly accounts for baseline in an optimal

ED

way. Missing responses not attributable to either death or an intervening non-lethal MACE will be handled using multiple imputation. Sensitivity analyses that conduct the imputations under

PT

a range of models that capture plausible deviations from the assumption, that data are

CE

missing at random, will be used to ensure a robust result. A separate risk benefit/analysis will be performed to study the treatment effect on intervening MACE events that do not prevent

AC

the successful performance of the 6MWD test. All randomized subjects will be followed and analyzed for both safety and efficacy endpoints on an intention to treat basis. Continuous variables will be summarized using descriptive statistics, and categorical variables will be summarized by frequencies and percentages. Sample Size Considerations: Based on results of the TAC-HFT study, a sample size of 250 in the randomized phase is expected to provide adequate power for the primary outcome, whereas the sample size was also based on the first hierarchical secondary outcome of non-

11

ACCEPTED MANUSCRIPT

inferiority analysis for all-cause mortality. The non-inferiority margin of 10% can be excluded from a 95% confidence interval with 90% power if the true survival rate in the cell treatment arm is 95% and the true survival rate in the sham arm is 96%. Details of the sample size

T

justification are provided in SUPPLEMENT 2.

IP

The trial sponsor is Biocardia Inc. (San Carlos, CA). The authors are solely responsible for the

CR

design and conduct of this study, all study analyses, the drafting and editing of the paper and its final contents.

US

Discussion

AN

The CardiAMP-HF trial is intended to expand and extend the results of the Phase II TAC-HFT study. The current study includes, to our knowledge, the first human use of a Cell Potency

M

Assay (CPA) to select patients who are likely to yield therapeutic cells and a high dose of BM

ED

MNC cells. It will test the hypothesis that intramyocardial delivery of high dose BM MNCs isolated with a point-of-care device will improve a clinical composite of 6MWD and MACE in

PT

NYHA functional class II-III ischemic heart failure patients with an LVEF of 20-40%. The use of a Cell Potency Assay (CPA) to select patients who are more likely to yield therapeutic cells,

CE

the high effective dose of BM MNCs that will be injected, and the point of care treatment approach are intended to maximize therapeutic efficacy, limit dose variability and improve

AC

treatment efficiency.

The CPA was developed to both enrich the enrollment of subjects that will have a greater yield of therapeutic cells and to reduce heterogeneity across subjects. The rationale for this approach is based on previous clinical trials where selected MNC subpopulations were administered in chronic myocardial ischemia patients28, 42 and were associated with improved outcomes. Reduced angina frequency was observed in the ACT-34 and RENEW trials that tested transendocardial injection of CD34+ cells in coronary artery disease patients with 12

ACCEPTED MANUSCRIPT

refractory chronic angina28, 37. Furthermore, in the FOCUS31, TIME47, and Late Time48 trials, certain cell sub-populations were associated with improved outcome following BM MNC treatment in patients with heart failure and recent or remote MI49. Age, and comorbidities were

T

also implicated as factors for responsiveness to cell therapy.

IP

Identifying biomarkers that are likely to predict therapeutic response to bone marrow-derived

CR

mononuclear cell therapy has been proposed in heart failure, recent acute myocardial infarction and critical limb ischemia. For example, the FOCUS trial demonstrated that

US

transendocardial injection of unselected BM MNCs in patients with ischemic cardiomyopathy was safe, but did not improve the co-primary outcomes of maximal oxygen consumption and

AN

perfusion31. However, left ventricular ejection fraction (LVEF) did improve in the BMMNC treated patients and those with increased LVEF had were younger than median age (<62

M

years) and had higher numbers of CD34+ and CD133+ cells in their BM31. Additionally,

ED

subjects who responded favorably had higher frequencies of B cells, CXCR4+ BM MNCs, fewer endothelial colony- forming cells and monocytes/macrophages in the bone marrow49.

PT

Demonstrating cardioregenerative responsiveness based on constituent bone marrow

CE

elements will be tested prospectively in the CardiAMP-HF trial. In an analysis of 63 biomarkers and cytokines in baseline plasma samples from myocardial infarction patients

AC

treated with BM MNCs or with cell free placebo, several markers were associated with favorable responses to cell therapy50. These bone marrow cell and plasma cytokines markers were also linked to endothelial progenitor cell function51. Similarly, expression of senescence genes was associated with increased mobilization efficiency and functional responsiveness of autologous CD133+ cells in no-revascularization option critical limb ischemia patients, in the Stem Cell Revascularization in Patients with Critical Limb Ischemia (SCRIPT-CLI) randomized trial52. The CPA approach used in the CardiAMP-HF trial also acts as a quality control on patient and dose variability, one of the great difficulties in autologous cell therapy trials to date. 13

ACCEPTED MANUSCRIPT

In addition to selection by CPA, CardiAMP-HF patients will be treated with a high cell dose. The target dose of 200 million MNCs is higher than most prior trials and will be delivered via transendocardial injection using a helical catheter previously shown to yield high acute cell retention within the myocardium compared to other methods53. Transendocardial and direct

T

intramyocardial cell delivery have been used extensively in pre-clinical models and human

IP

ischemic heart disease trials with an excellent safety profile54-56. This minimally invasive route

CR

offers the ability to deliver cells to all potential myocardial target segments as compared to intracoronary cell infusion, which relies on patent coronary arteries for delivery and can result

US

in microvascular obstruction due to cell plugging. Indeed, a meta-analysis of pre-clinical

AN

studies demonstrated that transendocardial delivery of mesenchymal stem cells in ischemia models improved cardiac outcomes to a greater extent than other delivery methods57.

M

The target cell dose in the CardiAMP Heart Failure Trial of 200 million BM MNCs is based on

ED

the dose used in the Phase II TAC-HFT trial32, and is a higher dose than previously has been used in randomized controlled trials of intramyocardial delivery of BM MNC. A BM MNC dose

PT

response was suggested by Silva et al. in a pre-clinical chronic ischemia model, where the

CE

~200 million cell dose showed greater efficacy than lower doses. Clinical trials using higher doses of BM MNCs also suggest a dose effect. Tse et al. reported in the PROTECT-CAD trial

AC

that intramyocardial injection of 42.0 ± 28.0 million MNCs improved exercise time, LVEF, and NYHA class25. Van Ramshorst et al. delivered 98 ± 6 million MNC showing a modest, but significant improvement in myocardial perfusion compared to placebo26. Perin et al. delivered 99.0 ± 5.6 million MNC and showed an improvement in LVEF, but failed to show an effect on other pre-specified primary endpoints, including left ventricular end systolic volume, maximal oxygen consumption, or reversibility on SPECT perfusion31. Most recently, in the REGENERATE-IHD trial, ischemic cardiomyopathy patients treated with 124.7 ± 89.2 million MNC experienced improvements in cardiac contractile function and heart failure symptoms58. 14

ACCEPTED MANUSCRIPT

To achieve a high “effective” cell dose, the CardiAMP study utilizes a helical injection catheter (“Helix” - see Figure 3) designed to maximize myocardial cell retention. Mitsutake et al. demonstrated acute retention of radiolabeled BM MNC was three-fold higher using the Helix catheter compared to straight needle injections53. In addition, transendocardial cell injection

T

using straight-needle catheters are associated with variable cell retention compared to

IP

intracoronary infusion59-61. The higher absolute BM MNC dose generated by the CardiAMP

CR

Cell Separation device, coupled with three fold higher estimated cell retention efficiency is expected to lead to an effective BM MNC dose up to five-fold higher than that delivered by

US

Mathur et al.58, which is the next highest cell dose delivered by intramuscular injection in a

AN

clinical trial to date.

In summary, the CardiAMP treatment “strategy” has several advantages. First, the autologous

M

cell therapy approach avoids the potential for immune rejection. While an off-the-shelf

ED

allogeneic approach has conceptual appeal, the inherent need for cell manipulation, cleanroom facilities for large scale cell production and storage, the shipment considerations, and

PT

contamination risk all present challenges. Second, the point-of-care approach to BM-MNC

CE

harvest, preparation and delivery utilized in this trial can easily be implemented within an existing interventional lab workflow. Relative accessibility, low cost, ease of procurement and

AC

simple cell preparation procedure, absence of immune response make autologous cell therapy an appealing adjunct to advanced heart failure therapy. CardiAMP-HF’s primary clinical composite endpoint uses 6MWD and MACE. The 6MWD has been shown to predict survival in patients with moderate to severe heart failure62-65. This simple to perform, patient-oriented, functional test was used as a primary efficacy endpoint in the Multicenter InSync Randomized Clinical Evaluation (MIRACLE) trial of cardiac resynchronization therapy (CRT), which enrolled a similar heart failure population to that

15

ACCEPTED MANUSCRIPT

proposed in CardiAMP-HF. In MIRACLE, six month 6MWD improved +39 meters versus +10 meters (CRT vs. control, p=0.001, n= 453 subjects randomized). This trial result contributed to the FDA decision to approve the Medtronic InSync CRT device66. In this study, MLWHFQ was also improved in the treatment group compared to control (–18.0 vs. –9.0 points, p= 0.001).

T

Notably, the relative differences in the point estimates between CRT and control for 6MWD

IP

and quality of life scores were smaller than the observed treatment benefit of BM MNCs

CR

observed in the aforementioned analysis of the TAC-HFT (CardiAMP Phase II) trial34. The

US

CardiAMP Phase III randomized trial is designed to confirm these treatment benefits. Conclusion: The CardiAMP pivotal trial will test whether transendocardial injection of a high

AN

effective dose autologous BM MNCs obtained from NYHA class II-III ischemic heart failure patients, who are selected by a novel bone marrow potency assay designed to assess the

M

likelihood of a therapeutic effect, improves ambulatory function, quality of life and survival.

ED

Success in this treatment approach may lead to a novel treatment option for those suffering with medically refractory ischemic heart failure.

PT

Acknowledgements: : The authors would like to acknowledge the contributions of Adrian

CE

Gee and April Durett of the Center for Cell & Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital in Houston; Maria Cabreira of the Stem Cell Center, Texas Heart

AC

Institute, Houston for their support of CardiAMP HF stem cell studies. In addition, Mary Jo Rizzo, Dr.Lavanya Bellumkonda, Dr.Lissa Sugeng Dr. Alexandra Lansky of Yale University are acknowledged for their contributions for the echocardiographic image analysis in the CardiAMP HF study. Finally, Michael Kolber, Cheryl Wong Po Foo, Andrew Mackenzie, and of Biocardia and Dr. Jerome Fleg of NHLBI are acknowledged for contributions toward developing the CardiAMP-HF trial protocol.

16

ACCEPTED MANUSCRIPT

Disclosures: The CardiAMP-HF trial is sponsored, in part, by Biocardia Inc. (San Carlos, CA). Authors ED and PA are employees of Biocardia Inc. The remaining authors are

AC

CE

PT

ED

M

AN

US

CR

IP

T

members of the executive steering committee for the CardiAMP-HF trial.

17

ACCEPTED MANUSCRIPT

REFERENCES Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S, Chiuve SE, Cushman M, Delling FN, Deo R, de Ferranti SD, Ferguson JF, Fornage M, Gillespie C, Isasi CR, Jimenez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Lutsey PL, Mackey JS, Matchar DB, Matsushita K, Mussolino ME, Nasir K, O'Flaherty M, Palaniappan LP, Pandey A, Pandey DK, Reeves MJ, Ritchey MD, Rodriguez CJ, Roth GA, Rosamond WD, Sampson UKA, Satou GM, Shah SH, Spartano NL, Tirschwell DL, Tsao CW, Voeks JH, Willey JZ, Wilkins JT, Wu JH, Alger HM, Wong SS, Muntner P. Heart disease and stroke statistics-2018 update: A report from the American Heart Association. Circulation. 2018;137:e67-e492 Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M, Magner M, Isner JM. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85:221-228 Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964-967 Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410:701-705 Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7:430-436 Zhang S, Zhang P, Guo J, Jia Z, Ma K, Liu Y, Zhou C, Li L. Enhanced cytoprotection and angiogenesis by bone marrow cell transplantation may contribute to improved ischemic myocardial function. European journal of Cardio-Thoracic Surgery. 2004;25:188-195 Shintani Y, Fukushima S, Varela-Carver A, Lee J, Coppen SR, Takahashi K, Brouilette SW, Yashiro K, Terracciano CM, Yacoub MH, Suzuki K. Donor cell-type specific paracrine effects of cell transplantation for post-infarction heart failure. Journal of molecular and cellular cardiology. 2009;47:288-295 Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, Noiseux N, Zhang L, Pratt RE, Ingwall JS, Dzau VJ. Paracrine action accounts for marked protection of ischemic heart by akt-modified mesenchymal stem cells. Nature medicine. 2005;11:367-368 Takahashi M, Li TS, Suzuki R, Kobayashi T, Ito H, Ikeda Y, Matsuzaki M, Hamano K. Cytokines produced by bone marrow cells can contribute to functional improvement of the infarcted heart by protecting cardiomyocytes from ischemic injury. American journal of physiology. Heart and circulatory physiology. 2006;291:H886-893 Kinnaird T, Stabile E, Burnett MS, Epstein SE. Bone-marrow-derived cells for enhancing collateral development: Mechanisms, animal data, and initial clinical experiences. Circ Res. 2004;95:354-363 Premer C, Blum A, Bellio MA, Schulman IH, Hurwitz BE, Parker M, Dermarkarian CR, DiFede DL, Balkan W, Khan A, Hare JM. Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine. 2015;2:467-475 Hatzistergos KE, Quevedo H, Oskouei BN, Hu Q, Feigenbaum GS, Margitich IS, Mazhari R, Boyle AJ, Zambrano JP, Rodriguez JE, Dulce R, Pattany PM, Valdes D, Revilla C, Heldman AW,

4.

8.

9.

10.

11.

12.

CR

PT

CE

7.

AC

6.

ED

M

5.

US

3.

AN

2.

IP

T

1.

18

ACCEPTED MANUSCRIPT

18.

19.

20.

21.

22.

23.

T

IP

CR

US

AN

M

17.

ED

16.

PT

15.

CE

14.

AC

13.

McNiece I, Hare JM. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circulation research. 2010;107:913-922 Fuchs S, Baffour R, Zhou YF, Shou M, Pierre A, Tio FO, Weissman NJ, Leon MB, Epstein SE, Kornowski R. Transendocardial delivery of autologous bone marrow enhances collateral perfusion and regional function in pigs with chronic experimental myocardial ischemia. J Am Coll Cardiol. 2001;37:1726-1732 Zhang S, Guo J, Zhang P, Liu Y, Jia Z, Ma K, Li W, Li L, Zhou C. Long-term effects of bone marrow mononuclear cell transplantation on left ventricular function and remodeling in rats. Life sciences. 2004;74:2853-2864 Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Tsutsumi Y, Ozono R, Masaki H, Mori Y, Iba O, Tateishi E, Kosaki A, Shintani S, Murohara T, Imaizumi T, Iwasaka T. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation. 2001;104:1046-1052 Tse HF, Siu CW, Zhu SG, Songyan L, Zhang QY, Lai WH, Kwong YL, Nicholls J, Lau CP. Paracrine effects of direct intramyocardial implantation of bone marrow derived cells to enhance neovascularization in chronic ischaemic myocardium. European journal of heart failure. 2007;9:747-753 Schneider C, Jaquet K, Geidel S, Rau T, Malisius R, Boczor S, Zienkiewicz T, Kuck KH, Krause K. Transplantation of bone marrow-derived stem cells improves myocardial diastolic function: Strain rate imaging in a model of hibernating myocardium. Journal of the American Society of Echocardiography : official publication of the American Society of Echocardiography. 2009;22:1180-1189 Silva GV, Fernandes MR, Cardoso CO, Sanz RR, Oliveira EM, Jimenez-Quevedo P, Lopez J, Angeli FS, Zheng Y, Willerson JT, Perin EC. A dosing study of bone marrow mononuclear cells for transendocardial injection in a pig model of chronic ischemic heart disease. Tex Heart Inst J. 2011;38:219-224 Leu S, Sun CK, Sheu JJ, Chang LT, Yuen CM, Yen CH, Chiang CH, Ko SF, Pei SN, Chua S, Youssef AA, Wu CJ, Yip HK. Autologous bone marrow cell implantation attenuates left ventricular remodeling and improves heart function in porcine myocardial infarction: An echocardiographic, six-month angiographic, and molecular-cellular study. International journal of cardiology. 2011;150:156-168 Pokushalov E, Romanov A, Chernyavsky A, Larionov P, Terekhov I, Artyomenko S, Poveshenko O, Kliver E, Shirokova N, Karaskov A, Dib N. Efficiency of intramyocardial injections of autologous bone marrow mononuclear cells in patients with ischemic heart failure: A randomized study. Journal of cardiovascular translational research. 2010;3:160-168 Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet. 2003;361:47-49 Fuchs S, Satler LF, Kornowski R, Okubagzi P, Weisz G, Baffour R, Waksman R, Weissman NJ, Cerqueira M, Leon MB, Epstein SE. Catheter-based autologous bone marrow myocardial injection in no-option patients with advanced coronary artery disease: A feasibility study. J Am Coll Cardiol. 2003;41:1721-1724 Beeres SL, Bax JJ, Zeppenfeld K, Dibbets-Schneider P, Stokkel MP, Fibbe WE, van der Wall EE, Schalij MJ, Atsma DE. Feasibility of trans-endocardial cell transplantation in chronic ischaemia. Heart. 2007;93:113-114 19

ACCEPTED MANUSCRIPT

28.

29.

T

IP

CR

AC

31.

CE

PT

30.

US

27.

AN

26.

M

25.

de la Fuente LM, Stertzer SH, Argentieri J, Penaloza E, Miano J, Koziner B, Bilos C, Altman PA. Transendocardial autologous bone marrow in chronic myocardial infarction using a helical needle catheter: 1-year follow-up in an open-label, nonrandomized, single-center pilot study (the tabmmi study). Am Heart J. 2007;154:79 e71-77 Tse HF, Thambar S, Kwong YL, Rowlings P, Bellamy G, McCrohon J, Thomas P, Bastian B, Chan JK, Lo G, Ho CL, Chan WS, Kwong RY, Parker A, Hauser TH, Chan J, Fong DY, Lau CP. Prospective randomized trial of direct endomyocardial implantation of bone marrow cells for treatment of severe coronary artery diseases (PROTECT-CAD trial). Eur Heart J. 2007 van Ramshorst J, Bax JJ, Beeres SL, Dibbets-Schneider P, Roes SD, Stokkel MP, de Roos A, Fibbe WE, Zwaginga JJ, Boersma E, Schalij MJ, Atsma DE. Intramyocardial bone marrow cell injection for chronic myocardial ischemia: A randomized controlled trial. JAMA. 2009;301:1997-2004 Losordo DW, Schatz RA, White CJ, Udelson JE, Veereshwarayya V, Durgin M, Poh KK, Weinstein R, Kearney M, Chaudhry M, Burg A, Eaton L, Heyd L, Thorne T, Shturman L, Hoffmeister P, Story K, Zak V, Dowling D, Traverse JH, Olson RE, Flanagan J, Sodano D, Murayama T, Kawamoto A, Kusano KF, Wollins J, Welt F, Shah P, Soukas P, Asahara T, Henry TD. Intramyocardial transplantation of autologous cd34+ stem cells for intractable angina: A phase i/iia double-blind, randomized controlled trial. Circulation. 2007;115:3165-3172 Losordo DW, Henry TD, Davidson C, Sup Lee J, Costa MA, Bass T, Mendelsohn F, Fortuin FD, Pepine CJ, Traverse JH, Amrani D, Ewenstein BM, Riedel N, Story K, Barker K, Povsic TJ, Harrington RA, Schatz RA. Intramyocardial, autologous cd34+ cell therapy for refractory angina. Circulation research. 2011;109:428-436 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;108:792-796 Perin EC, Silva GV, Henry TD, Cabreira-Hansen MG, Moore WH, Coulter SA, Herlihy JP, Fernandes MR, Cheong BY, Flamm SD, Traverse JH, Zheng Y, Smith D, Shaw S, Westbrook L, Olson R, Patel D, Gahremanpour A, Canales J, Vaughn WK, Willerson JT. A randomized study of transendocardial injection of autologous bone marrow mononuclear cells and cell function analysis in ischemic heart failure (FOCUS-HF). American heart journal. 2011;161:1078-1087 e1073 Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Silva GV, Lai D, Thomas JD, Kronenberg MW, Martin AD, Anderson RD, Traverse JH, Penn MS, Anwaruddin S, Hatzopoulos AK, Gee AP, Taylor DA, Cogle CR, Smith D, Westbrook L, Chen J, Handberg E, Olson RE, Geither C, Bowman S, Francescon J, Baraniuk S, Piller LB, Simpson LM, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD. Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: The focus-cctrn trial. JAMA. 2012;307:1717-1726 Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, Mushtaq M, Williams AR, Suncion VY, McNiece IK, Ghersin E, Soto V, Lopera G, Miki R, Willens H, Hendel R, Mitrani R, Pattany P, Feigenbaum G, Oskouei B, Byrnes J, Lowery MH, Sierra J, Pujol MV, Delgado C, Gonzalez PJ, Rodriguez JE, Bagno LL, Rouy D, Altman P, Foo CW, da Silva J, Anderson E, Schwarz R, Mendizabal A, Hare JM. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: The TAC-HFT randomized trial. JAMA. 2014;311:62-73

ED

24.

32.

20

ACCEPTED MANUSCRIPT

33.

34.

40.

41.

US

AN

AC

42.

M

39.

ED

38.

PT

37.

CE

36.

CR

IP

T

35.

Patel AN, Henry TD, Quyyumi AA, Schaer GL, Anderson RD, Toma C, East C, Remmers AE, Goodrich J, Desai AS, Recker D, DeMaria A. Ixmyelocel-t for patients with ischaemic heart failure: A prospective randomised double-blind trial. Lancet. 2016;387:2412-2421 Wong Po Foo C, Rouy D, Hare JM, Heldman AW, DiFede DL, McNiece IK, Mendizabal A, Kolber M, Altman P. The transendocardial autologous cells in ischemic heart failure trial bone marrow mononuclear cells (tac-hft-bmc) randomized placebo controlled blinded study. Regen Med. 2015;10:S169 de la Fuente LM, Stertzer SH, Argentieri J, Penaloza E, Koziner B, Rouy D, Altman PA. Transendocardial autologous bone marrow in myocardial infarction induced heart failure, two-year follow-up in an open-label phase i safety study (the tabmmi study). EuroIntervention : journal of EuroPCR in collaboration with the Working Group on Interventional Cardiology of the European Society of Cardiology. 2011;7:805-812 Fisher SA, Doree C, Mathur A, Taggart DP, Martin-Rendon E. Cochrane corner: Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Heart. 2017 Fisher SA, Doree C, Mathur A, Taggart DP, Martin-Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database Syst Rev. 2016;12:CD007888 Kawamoto A, Tkebuchava T, Yamaguchi J, Nishimura H, Yoon YS, Milliken C, Uchida S, Masuo O, Iwaguro H, Ma H, Hanley A, Silver M, Kearney M, Losordo DW, Isner JM, Asahara T. Intramyocardial transplantation of autologous endothelial progenitor cells for therapeutic neovascularization of myocardial ischemia. Circulation. 2003;107:461-468 Kawamoto A, Iwasaki H, Kusano K, Murayama T, Oyamada A, Silver M, Hulbert C, Gavin M, Hanley A, Ma H, Kearney M, Zak V, Asahara T, Losordo DW. Cd34-positive cells exhibit increased potency and safety for therapeutic neovascularization after myocardial infarction compared with total mononuclear cells. Circulation. 2006;114:2163-2169 Iwasaki H, Kawamoto A, Ishikawa M, Oyamada A, Nakamori S, Nishimura H, Sadamoto K, Horii M, Matsumoto T, Murasawa S, Shibata T, Suehiro S, Asahara T. Dose-dependent contribution of cd34-positive cell transplantation to concurrent vasculogenesis and cardiomyogenesis for functional regenerative recovery after myocardial infarction. Circulation. 2006;113:1311-1325 Henry TD, Schaer GL, Traverse JH, Povsic TJ, Davidson C, Lee JS, Costa MA, Bass T, Mendelsohn F, Fortuin FD, Pepine CJ, Patel AN, Riedel N, Junge C, Hunt A, Kereiakes DJ, White C, Harrington RA, Schatz RA, Losordo DW. Autologous CD34+ cell therapy for refractory angina: 2-year outcomes from the act34-cmi study. Cell transplantation. 2016;25:1701-1711 Povsic TJ, Henry TD, Traverse JH, Fortuin FD, Schaer GL, Kereiakes DJ, Schatz RA, Zeiher AM, White CJ, Stewart DJ, Jolicoeur EM, Bass T, Henderson DA, Dignacco P, Gu Z, Al-Khalidi HR, Junge C, Nada A, Hunt AS, Losordo DW. The renew trial: Efficacy and safety of intramyocardial autologous CD34(+) cell administration in patients with refractory angina. JACC. Cardiovascular interventions. 2016;9:1576-1585 Henry TD, Losordo DW, Traverse JH, Schatz RA, Jolicoeur EM, Schaer GL, Clare R, Chiswell K, White CJ, Fortuin FD, Kereiakes DJ, Zeiher AM, Sherman W, Hunt AS, Povsic TJ. Autologous cd34+ cell therapy improves exercise capacity, angina frequency and reduces mortality in nooption refractory angina: A patient-level pooled analysis of randomized double-blinded trials. European heart journal. 2018 Rector TS, Francis GS, Cohn JN. Patients' self assessment of their congestive heart failure. Part 1 patient perceived dysfunction and its poor correlation with maximal exercise tests. Heart Failure. 1987:192-196

43.

44.

21

ACCEPTED MANUSCRIPT

45.

49.

50.

AN

AC

CE

PT

48.

ED

M

47.

US

CR

IP

T

46.

Povsic TJ, Losordo DW, Story K, Junge CE, Schatz RA, Harrington RA, Henry TD. Incidence and clinical significance of cardiac biomarker elevation during stem cell mobilization, apheresis, and intramyocardial delivery: An analysis from ACT34-CMI. American heart journal. 2012;164:689-697 e683 Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD, Katus HA, Apple FS, Lindahl B, Morrow DA, Clemmensen PM, Johanson P, Hod H, Underwood R, Bax JJ, Bonow JJ, Pinto F, Gibbons RJ, Fox KA, Atar D, Newby LK, Galvani M, Hamm CW, Uretsky BF, Steg PG, Wijns W, Bassand JP, Menasche P, Ravkilde J, Ohman EM, Antman EM, Wallentin LC, Armstrong PW, Januzzi JL, Nieminen MS, Gheorghiade M, Filippatos G, Luepker RV, Fortmann SP, Rosamond WD, Levy D, Wood D, Smith SC, Hu D, Lopez-Sendon JL, Robertson RM, Weaver D, Tendera M, Bove AA, Parkhomenko AN, Vasilieva EJ, Mendis S, Baumgartner H, Ceconi C, Dean V, Deaton C, Fagard R, Funck-Brentano C, Hasdai D, Hoes A, Kirchhof P, Knuuti J, Kolh P, McDonagh T, Moulin C, Popescu BA, Reiner Z, Sechtem U, Sirnes PA, Torbicki A, Vahanian A, Windecker S, Morais J, Aguiar C, Almahmeed W, Arnar DO, Barili F, Bloch KD, Bolger AF, Botker HE, Bozkurt B, Bugiardini R, Cannon C, de Lemos J, Eberli FR, Escobar E, Hlatky M, James S, Kern KB, Moliterno DJ, Mueller C, Neskovic AN, Pieske BM, Schulman SP, Storey RF, Taubert KA, Vranckx P, Wagner DR. Third universal definition of myocardial infarction. Journal of the American College of Cardiology. 2012;60:1581-1598 Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG, Forder JR, Anderson RD, Hatzopoulos AK, Penn MS, Perin EC, Chambers J, Baran KW, Raveendran G, Lambert C, Lerman A, Simon DI, Vaughan DE, Lai D, Gee AP, Taylor DA, Cogle CR, Thomas JD, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Kappenman C, Westbrook L, Piller LB, Simpson LM, Baraniuk S, Loghin C, Aguilar D, Richman S, Zierold C, Spoon DB, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD. Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: The time randomized trial. JAMA. 2012:1-10 Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, Forder JR, Byrne BJ, Hatzopoulos AK, Penn MS, Perin EC, Baran KW, Chambers J, Lambert C, Raveendran G, Simon DI, Vaughan DE, Simpson LM, Gee AP, Taylor DA, Cogle CR, Thomas JD, Silva GV, Jorgenson BC, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Smith DX, Baraniuk S, Piller LB, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: The latetime randomized trial. JAMA. 2011;306:21102119 Taylor DA, Perin EC, Willerson JT, Zierold C, Resende M, Carlson M, Nestor B, Wise E, Orozco A, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Traverse JH, Cooke JP, Schutt RC, Bhatnagar A, Grant MB, Lai D, Johnstone BH, Sayre SL, Moye L, Ebert RF, Bolli R, Simari RD, Cogle CR. Identification of bone marrow cell subpopulations associated with improved functional outcomes in patients with chronic left ventricular dysfunction: An embedded cohort evaluation of the FOCUSCCTRN trial. Cell transplantation. 2016;25:1675-1687 Jokerst JV, Cauwenberghs N, Kuznetsova T, Haddad F, Sweeney T, Hou J, Rosenberg-Hasson Y, Zhao E, Schutt R, Bolli R, Traverse JH, Pepine CJ, Henry TD, Schulman IH, Moye L, Taylor DA, Yang PC. Circulating biomarkers to identify responders in cardiac cell therapy. Sci Rep. 2017;7:4419

22

ACCEPTED MANUSCRIPT

58.

59.

60.

61.

62.

63. 64.

T

IP

CR

US

57.

AN

56.

M

55.

ED

54.

PT

53.

CE

52.

Shahrivari M, Wise E, Resende M, Shuster JJ, Zhang J, Bolli R, Cooke JP, Hare JM, Henry TD, Khan A, Taylor DA, Traverse JH, Yang PC, Pepine CJ, Cogle CR. Peripheral blood cytokine levels after acute myocardial infarction: Il-1beta- and il-6-related impairment of bone marrow function. Circulation research. 2017;120:1947-1957 Raval AN, Schmuck EG, Tefera G, Leitzke C, Ark CV, Hei D, Centanni JM, de Silva R, Koch J, Chappell RG, Hematti P. Bilateral administration of autologous cd133+ cells in ambulatory patients with refractory critical limb ischemia: Lessons learned from a pilot randomized, double-blind, placebo-controlled trial. Cytotherapy. 2014;16:1720-1732 Mitsutake Y, Pyun WB, Rouy D, Foo CWP, Stertzer SH, Altman P, Ikeno F. Improvement of local cell delivery using helix transendocardial delivery catheter in a porcine heart. International heart journal. 2017;58:435-440 Raval AN. Therapeutic potential of adult progenitor cells in the management of chronic myocardial ischemia. American journal of cardiovascular drugs : drugs, devices, and other interventions. 2008;8:315-326 Sheng CC, Zhou L, Hao J. Current stem cell delivery methods for myocardial repair. Biomed Res Int. 2013;2013:547902 Perin EC, Lopez J. Methods of stem cell delivery in cardiac diseases. Nat Clin Pract Cardiovasc Med. 2006;3 Suppl 1:S110-113 Kanelidis AJ, Premer C, Lopez J, Balkan W, Hare JM. Route of delivery modulates the efficacy of mesenchymal stem cell therapy for myocardial infarction: A meta-analysis of preclinical studies and clinical trials. Circulation research. 2017;120:1139-1150 Choudhury T, Mozid A, Hamshere S, Yeo C, Pellaton C, Arnous S, Saunders N, Brookman P, Jain A, Locca D, Archbold A, Knight C, Wragg A, Davies C, Mills P, Parmar M, Rothman M, Choudry F, Jones DA, Agrawal S, Martin J, Mathur A. An exploratory randomized control study of combination cytokine and adult autologous bone marrow progenitor cell administration in patients with ischaemic cardiomyopathy: The regenerate-ihd clinical trial. European journal of heart failure. 2017;19:138-147 Perin EC, Silva GV, Assad JA, Vela D, Buja LM, Sousa AL, Litovsky S, Lin J, Vaughn WK, Coulter S, Fernandes MR, Willerson JT. Comparison of intracoronary and transendocardial delivery of allogeneic mesenchymal cells in a canine model of acute myocardial infarction. J Mol Cell Cardiol. 2008;44:486-495 Freyman T, Polin G, Osman H, Crary J, Lu M, Cheng L, Palasis M, Wilensky RL. A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction. European heart journal. 2006;27:1114-1122 van der Spoel TI, Vrijsen KR, Koudstaal S, Sluijter JP, Nijsen JF, de Jong HW, Hoefer IE, Cramer MJ, Doevendans PA, van Belle E, Chamuleau SA. Transendocardial cell injection is not superior to intracoronary infusion in a porcine model of ischaemic cardiomyopathy: A study on delivery efficiency. Journal of cellular and molecular medicine. 2012;16:2768-2776 Cahalin LP, Mathier MA, Semigran MJ, Dec GW, DiSalvo TG. The six-minute walk test predicts peak oxygen uptake and survival in patients with advanced heart failure. Chest. 1996;110:325332 Anand IS, Florea VG, Fisher L. Surrogate end points in heart failure. Journal of the American College of Cardiology. 2002;39:1414-1421 Arslan S, Erol MK, Gundogdu F, Sevimli S, Aksakal E, Senocak H, Alp N. Prognostic value of 6minute walk test in stable outpatients with heart failure. Texas Heart Institute journal. 2007;34:166-169

AC

51.

23

ACCEPTED MANUSCRIPT

66.

72.

73.

74.

75.

PT

CE

71.

AC

70.

ED

M

69.

AN

US

68.

CR

IP

67.

Shah MR, Hasselblad V, Gheorghiade M, Adams KF, Jr., Swedberg K, Califf RM, O'Connor CM. Prognostic usefulness of the six-minute walk in patients with advanced congestive heart failure secondary to ischemic or nonischemic cardiomyopathy. The American journal of cardiology. 2001;88:987-993 Abraham WT, Fisher WG, Smith AL, Delurgio DB, Leon AR, Loh E, Kocovic DZ, Packer M, Clavell AL, Hayes DL, Ellestad M, Trupp RJ, Underwood J, Pickering F, Truex C, McAtee P, Messenger J. Cardiac resynchronization in chronic heart failure. The New England journal of medicine. 2002;346:1845-1853 Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE, Jr., Drazner MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMurray JJ, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson LW, Tang WH, Tsai EJ, Wilkoff BL. 2013 accf/aha guideline for the management of heart failure: A report of the american college of cardiology foundation/american heart association task force on practice guidelines. Circulation. 2013;128:e240-327 Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW, George R, Lardo A. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The poseidon randomized trial. JAMA. 2012;308:2369-2379 Penn MS, Mendelsohn FO, Schaer GL, Sherman W, Farr M, Pastore J, Rouy D, Clemens R, Aras R, Losordo DW. An open-label dose escalation study to evaluate the safety of administration of nonviral stromal cell-derived factor-1 plasmid to treat symptomatic ischemic heart failure. Circulation research. 2013;112:816-825 Chung ES, Miller L, Patel AN, Anderson RD, Mendelsohn FO, Traverse J, Silver KH, Shin J, Ewald G, Farr MJ, Anwaruddin S, Plat F, Fisher SJ, AuWerter AT, Pastore JM, Aras R, Penn MS. Changes in ventricular remodelling and clinical status during the year following a single administration of stromal cell-derived factor-1 non-viral gene therapy in chronic ischaemic heart failure patients: The STOP-HF randomized phase ii trial. European heart journal. 2015;36:2228-2238 Cleland JG, Freemantle N, Erdmann E, Gras D, Kappenberger L, Tavazzi L, Daubert JC. Longterm mortality with cardiac resynchronization therapy in the cardiac resynchronization-heart failure (care-hf) trial. European journal of heart failure. 2012;14:628-634 Lechat P, Packer M, Chalon S, Cucherat M, Arab T, Boissel JP. Clinical effects of betaadrenergic blockade in chronic heart failure: A meta-analysis of double-blind, placebocontrolled, randomized trials. Circulation. 1998;98:1184-1191 Moss AJ, Hall WJ, Cannom DS, Klein H, Brown MW, Daubert JP, Estes NA, 3rd, Foster E, Greenberg H, Higgins SL, Pfeffer MA, Solomon SD, Wilber D, Zareba W. Cardiacresynchronization therapy for the prevention of heart-failure events. The New England journal of medicine. 2009;361:1329-1338 Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, Gilbert EM, Shusterman NH. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol heart failure study group. The New England journal of medicine. 1996;334:13491355 Saxon LA, Bristow MR, Boehmer J, Krueger S, Kass DA, De Marco T, Carson P, DiCarlo L, Feldman AM, Galle E, Ecklund F. Predictors of sudden cardiac death and appropriate shock in

T

65.

24

ACCEPTED MANUSCRIPT

T

IP

CR US AN M ED PT

78.

CE

77.

AC

76.

the comparison of medical therapy, pacing, and defibrillation in heart failure (COMPANION) trial. Circulation. 2006;114:2766-2772 Yusuf S, Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJ, Michelson EL, Olofsson B, Ostergren J. Effects of candesartan in patients with chronic heart failure and preserved leftventricular ejection fraction: The CHARM-PRESERVED trial. Lancet. 2003;362:777-781 Zannad F, McMurray JJ, Krum H, van Veldhuisen DJ, Swedberg K, Shi H, Vincent J, Pocock SJ, Pitt B. Eplerenone in patients with systolic heart failure and mild symptoms. The New England journal of medicine. 2011;364:11-21 Varela-Roman A, Grigorian L, Barge E, Bassante P, de la Pena MG, Gonzalez-Juanatey JR. Heart failure in patients with preserved and deteriorated left ventricular ejection fraction. Heart. 2005;91:489-494

25

ACCEPTED MANUSCRIPT TABLE 1. Key Inclusion and Exclusion Criteria

US

CR

IP

T

Inclusion Criteria 1. Age >21 and <90 years 2. New York Heart Association Class II or III 3. Chronic ischemic left ventricular dysfunction secondary to MI as defined by: - Previous MI (>6 months) documented by history that includes elevated cardiac biomarkers or ECG changes consistent with MI - Prior thrombolytic therapy, percutaneous coronary intervention or coronary artery bypass surgery 4. Left ventricular ejection fraction ≥20% and ≤40% assessed by echo core laboratory 5. All patients should receive stable, evidence based, medical and device therapy for heart failure or post 67 infarction left ventricular dysfunction (2013 ACC/AHA Heart Failure Guideline ) for at least 3 months prior to randomization. Pharmacotherapy: Stable pharmacologic therapy is defined as changes not exceeding halving or doubling the medications listed below. - Angiotensin converting enzyme inhibitors or angiotensin II receptor blockers or nitrate/hydralazine combination with investigator discretion after factoring in patient intolerances - Beta blockers approved for heart failure with investigator discretion after factoring in patient intolerances - Diuretics and aldosterone inhibitors Device therapy: cardiac resynchronization therapy (CRT) or CRT-Defibrillator therapy - CRT or CRT-D implant must be >3 months before randomization - Patients anticipated to be eligible for CRT or CRT-D within 6 months are excluded 6. Acceptable Cell Potency Assay Score as determined by the Cell Analysis Core Lab 7. Able to provide informed consent

AC

CE

PT

ED

M

AN

Exclusion Criteria 1. Severe lung disease (asthma or chronic obstructive pulmonary disease), orthopedic, muscular, or neurologic conditions that could limit the ability to perform the 6 Minute Walk Distance Test. 2. 6 Minute Walk Distance <100 meters or >450 meters 3. Ideal candidate for coronary revascularization 4. Patients anticipated to be eligible for CRT or CRT-D within 6 months are excluded 5. Severe mitral or tricuspid regurgitation as previously documented or measured by the echocardiographic core lab 6. Presence of a mechanical aortic valve or heart constrictive device 7. Presence of moderate or severe aortic valve stenosis or regurgitation 8. Acute coronary syndrome within 3 months 9. Life threatening arrhythmias or QTc interval >55ms on screening ECG 10. Baseline glomerular filtration rate <50 ml/min / 1.73m2 11. Hematological abnormality as evidenced by hematocrit <30%, white blood cell <4,500/µl or platelet values, <100,000/µl 12. Liver dysfunction, as evidenced by enzymes (AST and ALT) greater than three times the ULN) 13. Coagulopathy (INR ≥1.3) not due to a reversible cause (i.e., Coumadin). Patients on Coumadin will be withdrawn before the procedure and confirmed to have an INR <1.3. 14. Organ transplant recipient 15. 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 of skin, or cervical carcinoma 16. Serum positive for hepatitis B/C and/or HIV. 17. History of serious radiologic contrast allergy 18. Pregnant, nursing female or female of childbearing potential while not practicing effective contraceptive methods for at least 30 days prior to randomization

26

AC

CE

PT

ED

M

AN

US

CR

IP

T

ACCEPTED MANUSCRIPT

Figure 1. CardiAMP-HF trial workflow. ECG=electrocardiogram, 6MWD=six meter walk distance, HF=Heart Failure, BMMNC’s= Bone marrow mononuclear cells, PPM=permanent pacemaker, ICD=implanted cardiac defibrillator, MLWHFQ = Minnesota Living with Heart Failure Questionnaire, MACE=major adverse cardiac events

27

AN

US

CR

IP

T

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

Figure 2. CardiAMP-HF “Point of Care” Cell Isolation Process. From left to right, bone marrow aspirate is obtained from the posterior iliac crest and transferred into the cell separation chamber. The chamber is centrifuged and removed. The cell poor plasma layer is removed and the remaining nucleated product is drawn into a 10 mL syringe. This cell product is immediately distributed into 1 mL syringes for therapeutic transendocardial injections.

28

AN

US

CR

IP

T

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

Figure 3. Helix Transendocardial Delivery System. The Helix catheter has a helicalshaped needle with a distal lumen to administer the cell therapy and a more proximal lumen for injection of dilute contrast, which is used to verify transendocardial catheter engagement (left image). The Helix catheter is telescoped within the steerable Morph guide (right image). Both devices are positioned retrograde across the aortic valve into the left ventricle for transendocardial therapeutic injections.

29