throbber
PUBLISHED FEBRUARY 18, 2015
`Cardiac Fibrosis: New Treatments in Cardiovascular
`Medicine
`
`Kristin Reilly, PharmD
`Freelance Medical Writer
`East Windsor, New Jersey
`
`US Pharm. 2015;40(2):32-35.
`
`ABSTRACT: Almost 6 million people in the United States have heart
`failure. When heart failure develops, cardiac output decreases and
`compensatory mechanisms activate. One of these mechanisms is
`cardiac fibrosis, a scarring process that over time impacts cardiac
`structure and function. Historically, cardiac fibrosis has not been a focus
`for treatment; however, it is now believed that therapy directed at cardiac
`fibrosis could reduce the progression of heart failure and other
`cardiovascular diseases. Medications that target the renin-angiotensin
`system, transforming growth factor-beta, and endothelin are in various
`stages of development.
`
`Heart failure is a complex clinical syndrome in which structural or
`functional abnormalities impair the heart’s ability to fill with or pump
`blood. Affecting nearly 6 million people in the United States, heart
`1,2
`
`Lassen - Exhibit 1054, p. 1
`
`

`

`failure is the leading reason for hospitalization in patients aged 65 years
`and older, as well as a major cause of impaired quality of life and
`chronic disability.
`1-3
`
`Heart failure can result from systolic dysfunction, diastolic dysfunction,
`or both. The most common risk factors for developing heart failure
`1
`include coronary heart disease (often with myocardial infarction [MI]),
`hypertension, diabetes, and cardiomyopathy.
`3,4
`
`Heart failure is initiated by any event that impairs the heart’s ability to
`contract and/or relax, resulting in reduced cardiac output. As cardiac
`1
`output decreases, compensatory mechanisms activate to restore
`cardiac output through increased preload, tachycardia,
`vasoconstriction, ventricular hypertrophy, and remodeling. At the
`1,4
`cellular level, ventricular hypertrophy and remodeling are accompanied
`by cardiomyocyte hypertrophy, necrosis, apoptosis, fibroblast
`proliferation, and increased deposition of fibrous collagen, the last two
`of which are collectively termed cardiac fibrosis. This article will focus
`4
`on cardiac fibrosis, including its mediators, assessment, and potential
`treatments.
`
`Structure of the Heart Wall
`The heart wall is composed of three layers: the epicardium (outer layer),
`the myocardium (middle layer), and the endocardium (interior layer).
`5
`Fibrosis can occur in any layer and in various locations of the heart (i.e.,
`the four heart chambers and valves). The discussion in this article will
`3
`be limited to myocardial fibrosis.
`
`The myocardium contains a variety of cell types, including
`cardiomyocytes and fibroblasts. Fibroblasts are found within the
`6
`heart’s connective tissue and account for approximately two-thirds of
`cardiac cells.
`Fibroblasts are involved in many aspects of cardiac
`6-8
`function, including regulating the balance of the extracellular matrix
`(ECM), ECM remodeling, electrical activity, production of growth factors
`and cytokines, and intercellular signaling.
`6
`
`Lassen - Exhibit 1054, p. 2
`
`

`

`The structural component of the myocardium is the ECM. Collagen, a
`1
`fibrous protein found in ECM and connective tissue, is composed of
`amino acids.
`1
`
`Pathogenesis of Cardiac Fibrosis
`Ventricular remodeling, a natural compensatory process that precedes
`the development of heart failure symptoms, results in progressive
`changes in the structure and function of the heart, including cardiac
`hypertrophy, loss of cardiac muscle cells, and ECM alterations. Some
`1
`types of hypertrophy are accompanied by fibrosis.
`1
`
`Cardiac fibrosis occurs when fibroblasts are activated to
`myofibroblasts and produce elevated amounts of ECM proteins that
`form scar tissue and alter normal degradation of ECM (FIGURE 1).
`4,7,8
`Both processes lead to a buildup of collagen, which impacts both
`systolic and diastolic function.
`7,9
`
`Lassen - Exhibit 1054, p. 3
`
`

`

`Cardiac fibrosis, which is part of the normal aging process, is
`associated with many cardiovascular diseases, including heart failure,
`hypertension, and cardiomyopathies; it also is found in hearts that have
`been damaged by MI or radiation.
`Fibrosis progresses over time
`3,10-12
`and is accompanied by ongoing deterioration of heart function.
`13
`
`Types of Fibrosis
`Two forms of fibrosis—replacement fibrosis and reactive interstitial, or
`perivascular, fibrosis—have been identified. Replacement fibrosis
`10
`occurs in response to an injury causing cardiomyocyte death, as in the
`case of MI; a reparative response is activated in the heart, causing
`replacement of dead cells and formation of a collagen-based scar.
`10,14
`In reactive interstitial fibrosis, the cardiac interstitial space expands
`without significant cardiomyocyte loss.
`Reactive fibrosis allows the
`3,14
`heart to adapt to injury and retain its pressure-generating ability.
`14
`Pressure or volume overload, ischemia, and cardiomyopathies are
`examples of reactive fibrosis.
`14
`
`Mediators of Fibrosis
`Increases in various circulating hormones, cytokines, and proteins
`triggered by stress or injury contribute to fibroblast activation and
`differentiation and, ultimately, to cardiac fibrosis. Although many
`7,8
`substances have been identified as playing a part in this process,
`several studies suggest that the renin-angiotensin system (RAS),
`transforming growth factor (TGF)-beta, and endothelin (ET) are key
`elements in the cascade. These elements will be the focus of this
`discussion.
`8
`
`The RAS regulates the production and activity of fibroblasts. When the
`11
`heart is injured, macrophages and fibroblasts produce renin and ACE,
`which in turn generate angiotensin II (Ang II). Ang II interacts with Ang
`9
`II receptor type 1 (AT ) receptors, which promote hypertrophy, stimulate
`1
`
`Lassen - Exhibit 1054, p. 4
`
`

`

`In addition,
`fibroblast proliferation, and increase collagen synthesis.
`9,15
`Ang II suppresses collagenase (an enzyme that breaks down collagen),
`which may lead to increased collagen accumulation and fibrosis.
`13
`
`Aldosterone has also been identified as a mediator of fibrosis. It affects
`fibroblast proliferation and collagen deposition in the ECM, heightening
`expression of cytokines and chemokines, signaling macrophages, and
`activating cardiomyocyte fibrogenic signals.
`1,9,12,13
`
`TGF-beta is a cytokine in the heart that is activated by cardiac injury,
`generation of reactive oxygen species, Ang II, high glucose, altered pH,
`and certain proteases. Once activated, TGF-beta increases ECM
`4,9
`production and decreases ECM breakdown.
`4,9
`
`It has
`ET is a protein that is released by endothelial cells in the heart.
`8
`been found to increase fibroblast differentiation into myofibroblasts
`and the production of ECM.
`8
`
`Assessment of Fibrosis
`Historically, the only measures of cardiac fibrosis have been
`echocardiograms that indirectly measure left ventricle mass and
`endomyocardial biopsies that measure collagen volume fraction
`(CVF). Newer techniques involve laboratory assessment of
`11
`biomarkers and cardiac imaging.
`11
`
`Several biomarkers for fibrosis have been identified, most of which are
`focused on ECM structure. Researchers can measure biomarkers of
`11
`the synthesis and degradation of collagen types I and III, the main
`components of ECM. Collagen types I and III are synthesized as
`11
`procollagens, which are then processed into mature collagen molecules
`by a peptidase cleaving their propeptide domain.
`During collagen
`6,11
`synthesis, propeptides from the amino-terminals (PINP and PIIINP) or
`carboxy-terminals (PICP and PIIICP) of collagen types I and III are
`released and measured as biomarkers. During collagen degradation,
`6
`
`Lassen - Exhibit 1054, p. 5
`
`

`

`telopeptides in the amino-terminals (NITP, NIIITP) or carboxy-terminals
`(CITP, CIIITP) of collagen types I and III are cleaved and act as
`biomarkers.
`6
`
`Biomarkers may be used to identify fibrosis before symptoms of
`disease are present, as well as to assess the efficacy of medications.
`Biomarkers are commonly used as endpoints in clinical trials.
`11
`
`MRI is useful for visualizing certain types of fibrosis. Contrast-
`11
`enhanced cardiac MRI can readily visualize “patchy,” or regional,
`myocardial fibrosis from MI or infiltration.
`Diffuse fibrosis is much
`11,12
`more difficult to visualize, and research into enhanced MRI techniques
`may prove beneficial in identifying this and other forms of fibrosis.
`11
`Other imaging techniques that have shown promise in identifying
`cardiac fibrosis include single photon emission CT and positron
`emission tomography scanning; more studies are required, however.
`11
`
`Potential Treatments
`For certain conditions that cause fibrosis, such as volume or pressure
`overload, the best treatment is to prevent fibrosis by controlling risk
`factors.
`For patients in whom prevention is not possible or has failed,
`9,14
`existing drugs and new chemical entities are in various stages of
`development; these products may have a significant effect on the
`management of many cardiovascular diseases.
`8,11
`
`Agents That Impact the RAS: As stated previously, the RAS plays a
`central role in fibroblast activation; as such, it is an important target for
`drug therapy. A significant amount of research has been conducted
`11
`on how drugs that modify this system affect cardiac fibrosis; specific
`information is provided in TABLE 1.
`
`Lassen - Exhibit 1054, p. 6
`
`

`

`Several antihypertensive classes, including beta-blockers and calcium
`channel blockers, have shown efficacy in reducing fibrosis in animals;
`however, results in humans have been inconsistent.
`RAS inhibitors,
`8,11
`such as ACE inhibitors, Ang II receptor blockers (ARBs), and
`aldosterone antagonists, have demonstrated positive results in animals
`and humans. One study of lisinopril versus hydrochlorothiazide
`11
`(HCTZ) in patients with hypertension, left ventricular hypertrophy, and
`left ventricular diastolic dysfunction found that lisinopril decreased CVF
`significantly compared with HCTZ. Blood pressure (BP) was
`13
`controlled in both treatment groups, but the effect on fibrosis differed.
`13
`
`ARBs also are effective for reducing fibrosis. Losartan and olmesartan
`11
`have favorable animal data, and candesartan and losartan have reduced
`fibrosis biomarkers in humans.
`In a 1-year study of losartan versus
`8,11
`amlodipine, losartan significantly decreased fibrosis, whereas
`amlodipine did not; both drugs affected BP similarly, however. These
`11
`studies of lisinopril and losartan show that treatments can impact
`fibrosis and hypertension independently of each other.
`11
`
`Lassen - Exhibit 1054, p. 7
`
`

`

`Unlike ACE inhibitors and ARBs, aldosterone antagonists directly block
`aldosterone, which may be a more effective way of reducing its
`profibrotic effects. Spironolactone and eplerenone have been shown
`11
`to reduce myocardial fibrosis in animals and humans, though results in
`humans are mixed.
`2,11
`
`Vaccines with anti–Ang II effects are being researched in animals and
`humans, mostly for hypertension.
`One vaccine effectively decreased
`11,16
`cardiac fibrosis in immunized mice; Ang II signaling was inhibited, and
`anti–Ang II antibodies increased.
`11
`
`In
`The RAS is a complex system with two counterbalancing axes.
`11,15
`addition to the familiar ACE/Ang II/AT axis, an ACE2/Ang-(1-7)/Mas
`1
`receptor axis has been identified.
`ACE2 hydrolyzes Ang II into
`11,15
`Ang-(1-7), and Mas is a protein receptor for Ang-(1-7).
`The
`11,15
`ACE2/Ang-(1-7)/Mas receptor axis has shown antifibrogenic and
`antiproliferative effects in various organs, including the heart. Several
`15
`animal studies have provided strong evidence that administration of
`Ang-(1-7) and overexpression of ACE2 can reduce cardiac fibrosis.
`15
`While therapeutic agents for these areas have not yet been developed,
`the foundation for future research has been built.
`15
`
`TGF-beta Inhibitors: As previously discussed, TGF-beta plays a central
`role in activating cardiac fibrosis, and inhibiting its actions could have
`profound effects on reducing fibrosis. Several approaches to inhibiting
`4
`TGF-beta are being researched; two agents, pirfenidone and tranilast,
`are the furthest developed. Although the exact mechanisms of action
`are not completely understood, these drugs appear to inhibit TGF-beta
`and other growth factors.
`4
`
`Pirfenidone, an oral medication, was approved in October 2014 for the
`treatment of idiopathic pulmonary fibrosis.
`In animal studies,
`17
`pirfenidone has shown efficacy in reducing cardiac fibrosis, decreasing
`left atrial remodeling, and reducing diastolic stiffness, but not restoring
`cardiac contractility.
`4
`
`Lassen - Exhibit 1054, p. 8
`
`

`

`Tranilast has been used in Japan for more than 20 years to treat
`asthma, allergic rhinitis, and atopic dermatitis.
`In animal studies,
`4,18
`tranilast reduced cardiac fibrosis without affecting BP, suggesting a
`direct effect on fibrosis.
`In a human study examining prevention of
`4
`restenosis after percutaneous coronary intervention, however,
`quantitative measures of efficacy were not found; moreover, several
`laboratory abnormalities were detected that could impact the use of
`tranilast, including increased bilirubin, liver enzymes exceeding three
`times the upper limit of normal, and serum creatinine increases of 50%.
`Research is being conducted on new compounds that could overcome
`some of these potential safety concerns.
`4
`
`ET Inhibitors: Currently, several ET receptor inhibitors are approved in
`the U.S. for the treatment of pulmonary hypertension, including the dual
`ET subtype A/ET subtype B (ETA/ETB) inhibitors bosentan and
`macitentan and the ETA inhibitor ambrisentan.
`It is postulated that
`8,19-21
`it may be necessary to target dual ETA/ETB inhibition, since both
`receptors impact fibrosis.
`In animal studies, bosentan has been
`22
`demonstrated to inhibit ECM formation, decrease collagen synthesis,
`and increase collagenase suppression. Studies examining its use in
`22
`fibrosis are ongoing.
`8
`
`Drugs Impacting Other Mediators of Fibrosis: Histone deacetylases
`(HDACs) are enzymes that play a key role in regulating gene
`transcription throughout the body. HDACs may be linked to the
`7
`signaling of some cellular molecules that have an effect on cardiac
`fibrosis, as well as inflammation. Animal studies have revealed that
`7
`HDAC inhibitors can stop, or even reverse, cardiac fibrosis. One
`7
`inhibitor has positive results in reducing cardiac fibrosis, as well as
`levels of Ang II receptors and TGF-beta. Although a great deal of
`7
`research is needed, HDAC inhibitors hold promise as future treatment.
`
`7
`
`Ivabradine is an oral medication currently available outside the U.S. that
`provides selective heart rate reduction by inhibiting the f-channel of the
`sinoatrial node.
`In August 2014, the FDA granted fast-track
`23
`
`Lassen - Exhibit 1054, p. 9
`
`

`

`designation for this compound in the treatment of chronic heart
`failure.
`In animal models, ivabradine effectively reduced fibrosis and
`23
`circulating Ang II and aldosterone levels.
`11
`
`Additional agents, including diltiazem, tadalafil, isosorbide dinitrate and
`hydralazine, erythropoietin, cyclosporine, thalidomide, and anti-
`inflammatory drugs impacting cytokines (e.g., tumor necrosis factor-
`alpha, interleukin [IL]-1, and IL-6), are being evaluated for fibrosis.
`11
`Transplantation of a variety of stem cells following MI has been
`demonstrated to decrease cardiac fibrosis and cardiac muscle
`apoptosis.
`11,14
`
`Conclusion
`Cardiac fibrosis is believed to be the final pathway leading to heart
`failure, which is an extremely common syndrome in the U.S. Much of
`the pathophysiology of cardiac fibrosis is known. Many drug treatments
`have been studied and seem promising, but data in humans are both
`limited and mixed. Although more studies must be conducted to
`evaluate the safety and efficacy of possible treatments for cardiac
`fibrosis, there is great hope for the future.
`
`REFERENCES
`1. Parker RB, Nappi JM, Cavallari LH. Chronic heart failure. In: DiPiro JT, Talbert RL, Yee GC, et
`al, eds. Pharmacotherapy: A Pathophysiologic Approach. 9th ed. New York, NY: McGraw-Hill
`Education; 2014.
`2. Rich MW. Heart failure. In: Halter JB, Ouslander JG, Tinetti ME, et al, eds. Hazzard’s Geriatric
`Medicine and Gerontology. 6th ed. McGraw-Hill; 2009.
`3. Biernacka A, Frangogiannis NG. Aging and cardiac fibrosis. Aging Dis. 2011;2:158-173.
`4. Edgley AJ, Krum H, Kelly DJ. Targeting fibrosis for the treatment of heart failure: a role for
`transforming growth factor-beta. Cardiovasc Ther. 2012;30:e30-e40.
`5. Mescher AL. Junqueira’s Basic Histology: Text and Atlas. 13th ed. New York, NY: McGraw-Hill;
`2013:212-233.
`6. Fan D, Takawale A, Lee J, Kassiri Z. Cardiac fibroblasts, fibrosis and extracellular matrix
`remodeling in heart disease. Fibrogenesis Tissue Repair. 2012;5:15.
`7. Tao H, Shi KH, Yang JJ, et al. Histone deacetylases in cardiac fibrosis: current perspectives
`for therapy. Cell Signal. 2014;26:521-527.
`8. Leask A. Potential therapeutic targets for cardiac fibrosis: TGFbeta, angiotensin, endothelin,
`CCN2, and PDGF, partners in fibroblast activation. Circ Res. 2010;106:1675-1680.
`
`Lassen - Exhibit 1054, p. 10
`
`

`

`9. Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci.
`2014;71:549-574.
`10. Krenning G, Zeisberg EM, Kalluri R. The origin of fibroblasts and mechanism of cardiac
`fibrosis. J Cell Physiol. 2010;225:631-637.
`11. Roubille F, Busseuil D, Merlet N, et al. Investigational drugs targeting cardiac fibrosis. Expert
`Rev Cardiovasc Ther. 2014;12:111-125.
`12. Bashore TM, Granger CB, Jackson K, Patel MR. Heart disease. In: Papadakis MA, McPhee
`SJ, eds. Current Medical Diagnosis & Treatment 2015. New York, NY: McGraw-Hill Education;
`2015.
`13. Brilla CG, Funck RC, Rupp H. Lisinopril-mediated regression of myocardial fibrosis in
`patients with hypertensive heart disease. Circulation. 2000;102:1388-1393.
`14. Elnakish MT, Kuppusamy P, Khan M. Stem cell transplantation as a therapy for cardiac
`fibrosis. J Pathol. 2013;229:347-354.
`15. Simões e Silva AC, Silveira KD, Ferreira AJ, Teixeira MM. ACE2, angiotensin-(1-7) and Mas
`receptor axis in inflammation and fibrosis. Br J Pharmacol. 2013;169:477-492.
`16. Bairwa M, Pilania M, Gupta V, Yadav K. Hypertension vaccine may be a boon to millions in
`developing world. Hum Vaccin Immunother. 2014;30:708-713.
`17. FDA. FDA approves Esbriet to treat idiopathic pulmonary fibrosis.
`www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm418991.htm. Accessed
`October 27, 2014.
`18. Kissei Pharmaceutical Co. Kissei entered into an agreement with Nuon Therapeutics, Inc on
`Tranilast. www.kissei.co.jp/e_contents/relation/2007/20070723.html. Accessed October 22,
`2014.
`19. Tracleer (bosentan) product information. South San Francisco, CA: Actelion
`Pharmaceuticals US, Inc; October 2012.
`20. Letairis (ambrisentan) product information. Foster City, CA: Gilead Sciences, Inc; May 2014.
`21. Opsumit (macitentan) product information. South San Francisco, CA: Actelion
`Pharmaceuticals US, Inc; October 2013.
`22. Clozel M, Salloukh H. Role of endothelin in fibrosis and anti-fibrotic potential of bosentan.
`Ann Med. 2005;37:2-12.
`23. Amgen. FDA grants Amgen priority review designation for ivabradine for the treatment of
`chronic heart failure. www.amgen.com/media/media_pr_detail.jsp?releaseID=1961392.
`Accessed October 22, 2014.
`
`To comment on this article, contact rdavidson@uspharmacist.com.
`
`We recommend
`
`A Pharmacist’s Guide for Systolic Heart Failure
`Angela R. Thomason et al., US Pharmacist,
`2006
`
`Managing Hypertrophic Cardiomyopathy
`
`Protein Kinase Cε-Calcineurin Cosignaling
`Downstream of Toll-Like Receptor 4
`Downregulates Fibrosis and Induces Wound
`Healing Gene Expression in Cardiac
`Myofibroblasts
`Mol Cell Biol, 2014
`
`Lassen - Exhibit 1054, p. 11
`
`

`

`Manouchkathe Cassagnol et al., US
`Pharmacist, 2016
`
`Aortic Stenosis: Evidence Supports Surgery
`Mary Ann E. Zagaria et al., US Pharmacist,
`2011
`
`Management of Arrhythmias, Part II:
`Ventricular Arrhythmias
`Mindi S. Miller et. al., US Pharmacist, 2006
`
`Rate Control in Atrial Fibrillation
`Mary Ann E. Zagaria et al., US Pharmacist,
`2007
`
`Powered by
`
`Deficiency of Cardiomyocyte-specific
`MicroRNA-378 Contributes to the
`Development of Cardiac Fibrosis Involving a
`Transforming Growth Factor β (TGFβ1)
`-dependent Paracrine Mechanism
`Raghu S. Nagalingam et al., Journal of
`Biological Chemistry, 2014
`
`Cell-specific ablation of Hsp47 defines the
`collagen producing cells in the injured heart
`
`Hadi Khalil et al., JCI Insight, 2019
`
`191 Role of mir-214 in angiotensin ii induced
`hypertensive heart disease
`Eilidh McGinnigle et al., Heart, 2017
`
`Myocyte-Derived Hsp90 Modulates Collagen
`Upregulation via Biphasic Activation of
`STAT-3 in Fibroblasts during Cardiac
`Hypertrophy
`Mol Cell Biol, 2017
`
`Copyright © 2000 - 2019 Jobson Medical Information LLC unless otherwise noted. All
`rights reserved. Reproduction in whole or in part without permission is prohibited.
`
`Lassen - Exhibit 1054, p. 12
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket