E-Mail Us Close
Please note that this email should only be used for feedback and comments specifically related to this particular medical policy.
  
Horizon BCBSNJ
Uniform Medical Policy ManualSection:Treatment
Policy Number:148
Effective Date: 06/14/2019
Original Policy Date:02/24/2015
Last Review Date:02/11/2020
Date Published to Web: 03/13/2019
Subject:
Bioengineered Skin and Soft Tissue Substitutes

Description:
_______________________________________________________________________________________

IMPORTANT NOTE:

The purpose of this policy is to provide general information applicable to the administration of health benefits that Horizon Blue Cross Blue Shield of New Jersey and Horizon Healthcare of New Jersey, Inc. (collectively “Horizon BCBSNJ”) insures or administers. If the member’s contract benefits differ from the medical policy, the contract prevails. Although a service, supply or procedure may be medically necessary, it may be subject to limitations and/or exclusions under a member’s benefit plan. If a service, supply or procedure is not covered and the member proceeds to obtain the service, supply or procedure, the member may be responsible for the cost. Decisions regarding treatment and treatment plans are the responsibility of the physician. This policy is not intended to direct the course of clinical care a physician provides to a member, and it does not replace a physician’s independent professional clinical judgment or duty to exercise special knowledge and skill in the treatment of Horizon BCBSNJ members. Horizon BCBSNJ is not responsible for, does not provide, and does not hold itself out as a provider of medical care. The physician remains responsible for the quality and type of health care services provided to a Horizon BCBSNJ member.

Horizon BCBSNJ medical policies do not constitute medical advice, authorization, certification, approval, explanation of benefits, offer of coverage, contract or guarantee of payment.

__________________________________________________________________________________________________________________________

Bioengineered skin and soft tissue substitutes may be derived from human tissue (autologous or allogeneic), nonhuman tissue (xenographic), synthetic materials, or a composite of these materials. Bioengineered skin and soft tissue substitutes are being evaluated for a variety of conditions, including breast reconstruction and healing lower-extremity ulcers and severe burns. Acellular dermal matrix (ADM) products are also being evaluated for soft tissue repair.

PopulationsInterventionsComparatorsOutcomes
Individuals:
  • Who are undergoing breast reconstruction
Interventions of interest are:
  • Allogeneic acellular dermal matrix products
Comparators of interest are:
  • Breast reconstruction without an acellular dermal matrix product
Relevant outcomes include:
  • Symptoms
  • Morbid events
  • Functional outcomes
  • Quality of life
  • Treatment-related morbidity
Individuals:
  • Who are undergoing tendon repair
Interventions of interest are:
  • Graftjacket
Comparators of interest are:
  • Surgical repair alone
Relevant outcomes include:
  • Symptoms
  • Morbid events
  • Functional outcomes
  • Quality of life
  • Treatment-related morbidity
Individuals:
  • Who are undergoing surgical repair of hernias or parastomal reinforcement
Interventions of interest are:
  • Acellular collagen-based scaffolds
Comparators of interest are:
  • Surgical repair alone
  • Standard surgical mesh
Relevant outcomes include:
  • Symptoms
  • Morbid events
  • Functional outcomes
  • Quality of life
  • Treatment-related morbidity
Individuals:
  • With diabetic lower- extremity ulcers
Interventions of interest are:
  • Apligraf, Dermagraft, AlloPatch, or Integra
Comparators of interest are:
  • Standard wound care
Relevant outcomes include:
  • Disease-specific survival
  • Symptoms
  • Change in disease status
  • Morbid events
  • Quality of life
Individuals:
  • With diabetic lower- extremity ulcers
Interventions of interest are:
  • Acellular dermal matrix products other than Apligraf, Dermagraft, AlloPatch, or Integra
Comparators of interest are:
  • Standard wound care
Relevant outcomes include:
  • Disease-specific survival
  • Symptoms
  • Change in disease status
  • Morbid events
  • Quality of life
Individuals:
  • With lower-extremity ulcers due to venous insufficiency
Interventions of interest are:
  • Apligraf and Oasis Wound Matrix
Comparators of interest are:
  • Standard wound care
Relevant outcomes include:
  • Disease-specific survival
  • Symptoms
  • Change in disease status
  • Morbid events
  • Quality of life
Individuals:
  • With lower-extremity ulcers due to venous insufficiency
Interventions of interest are:
  • Bioengineered skin substitutes other than Apligraf and Oasis Wound Matrix
Comparators of interest are:
  • Standard wound care
Relevant outcomes include:
  • Disease-specific survival
  • Symptoms
  • Change in disease status
  • Morbid events
  • Quality of life
Individuals:
  • With dystrophic epidermolysis bullosa
Interventions of interest are:
  • Bioengineered skin substitutes (ie, OrCel)
Comparators of interest are:
  • Standard wound care
Relevant outcomes include:
  • Symptoms
  • Change in disease status
  • Morbid events
  • Quality of life
Individuals:
  • With deep dermal burns
Interventions of interest are:
  • Bioengineered skin substitutes (ie, Epicel, Integra Dermal Regeneration Template)
Comparators of interest are:
  • Standard wound care
Relevant outcomes include:
  • Disease-specific survival
  • Symptoms
  • Morbid events
  • Functional outcomes
  • Quality of life
  • Treatment-related morbidity

BACKGROUND

Skin and Soft Tissue Substitutes

Bioengineered skin and soft tissue substitutes may be either acellular or cellular. Acellular products (eg, dermis with cellular material removed) contain a matrix or scaffold composed of materials such as collagen, hyaluronic acid, and fibronectin. Acellular dermal matrix (ADM) products can differ in a number of ways, including as species source (human, bovine, porcine), tissue source (eg dermis, pericardium, intestinal mucosa), additives (eg antibiotics, surfactants), hydration (wet, freeze-dried), and required preparation (multiple rinses, rehydration).

Cellular products contain living cells such as fibroblasts and keratinocytes within a matrix. The cells contained within the matrix may be autologous, allogeneic, or derived from other species (eg, bovine, porcine). Skin substitutes may also be composed of dermal cells, epidermal cells, or a combination of dermal and epidermal cells, and may provide growth factors to stimulate healing. Bioengineered skin substitutes can be used as either temporary or permanent wound coverings.

Applications

There are a large number of potential applications for artificial skin and soft tissue products. One large category is nonhealing wounds, which potentially encompasses diabetic neuropathic ulcers, vascular insufficiency ulcers, and pressure ulcers. A substantial minority of such wounds do not heal adequately with standard wound care, leading to prolonged morbidity and increased risk of mortality. For example, nonhealing lower-extremity wounds represent an ongoing risk for infection, sepsis, limb amputation, and death. Bioengineered skin and soft tissue substitutes have the potential to improve rates of healing and reduce secondary complications.

Other situations in which bioengineered skin products might substitute for living skin grafts include certain postsurgical states (eg, breast reconstruction) in which skin coverage is inadequate for the procedure performed, or for surgical wounds in patients with compromised ability to heal. Second- and third-degree burns are another indication in which artificial skin products may substitute for auto- or allografts. Certain primary dermatologic conditions that involve large areas of skin breakdown (eg, bullous diseases) may also be conditions in which artificial skin products can be considered as substitutes for skin grafts. ADM products are also being evaluated in the repair of other soft tissues including rotator cuff repair, following oral and facial surgery, hernias, and other conditions.

Regulatory Status

A large number of artificial skin products are commercially available or in development. The following summary of commercially available skin substitutes describes those products that have substantial relevant evidence on efficacy.

ADM Products

Allograft ADM products derived from donated human skin tissue are supplied by tissue banks compliant with standards of the American Association of Tissue Banks and U.S. Food and Drug Administration (FDA) guidelines. The processing removes the cellular components (ie, epidermis, all viable dermal cells) that can lead to rejection and infection. ADM products from human skin tissue are regarded as minimally processed and not significantly changed in structure from the natural material; FDA classifies ADM products as banked human tissue and, therefore, not requiring FDA approval.

·         AlloDerm® (LifeCell Corp.) is an ADM (allograft) tissue-replacement product created from native human skin and processed so that the basement membrane and cellular matrix remain intact. Originally, AlloDerm® required refrigeration and rehydration before use. It is currently available in a ready-to-use product stored at room temperature. An injectable micronized form of AlloDerm® (Cymetra) is available.

·         Cortiva® (previously marketed as AlloMax™ Surgical Graft and before that NeoForm™) is an acellular non-cross-linked human dermis allograft.

·         AlloPatch® (Musculoskeletal Transplant Foundation) is an acellular human dermis allograft derived from the reticular layer of the dermis and marketed for wound care. This product is also marketed as FlexHD® for postmastectomy breast reconstruction.

·         FlexHD® and the newer formulation FlexHD® Pliable™ (Musculoskeletal Transplant Foundation) are acellular hydrated reticular dermis allograft derived from donated human skin.

·         DermACELL™ (LifeNet Health) is an allogeneic ADM processed with proprietary technologies MATRACELL® and PRESERVON®.

·         DermaMatrix™ (Synthes) is a freeze-dried ADM derived from donated human skin tissue. DermaMatrix Acellular Dermis is processed by the Musculoskeletal Transplant Foundation.

·         DermaPure™ (Tissue Regenix Wound Care) is a single-layer decellularized human dermal allograft for the treatment of acute and chronic wounds.

·         Graftjacket® Regenerative Tissue Matrix (also called Graftjacket Skin Substitute; KCI) is an acellular regenerative tissue matrix that has been processed from human skin supplied from U.S. tissue banks. The allograft is minimally processed to remove the epidermal and dermal cells while preserving dermal structure. Graftjacket Xpress® is an injectable product.

FDA product codes: FTM, OXF.

Xenogenic Products

Cytal™ (previously called MatriStem®) Wound Matrix, Multilayer Wound Matrix, Pelvic Floor Matrix, MicroMatrix, and Burn Matrix (all manufactured by ACell) are composed of porcine-derived urinary bladder matrix.

Helicoll (Encol) is an acellular collagen matrix derived from bovine dermis. In 2004, it was cleared for marketing by FDA through the 510(k) process for topical wound management that includes partial and full-thickness wounds, pressure ulcers, venous ulcers, chronic vascular ulcers, diabetic ulcers, trauma wounds (eg, abrasions, lacerations, second-degree bums, skin tears), and surgical wounds including donor sites/grafts.

Keramatrix® (Keraplast Research) is an open-cell foam comprised of freeze-dried keratin that is derived from acellular animal protein. In 2009, it was cleared for marketing by FDA through the 510(k) process under the name of Keratec. The wound dressings are indicated in the management of the following types of dry, light, and moderately exudating partial and full-thickness wounds: pressure (stage I-IV) and venous stasis ulcers, ulcers caused by mixed vascular etiologies, diabetic ulcers, donor sites, and grafts.

Kerecis™ Omega3 Wound (Kerecis) is an ADM derived from fish skin. It has a high content of omega 3 fatty acids and is intended for use in burn wounds, chronic wounds, and other applications.

Permacol™ (Covidien) is xenogeneic and composed of cross-linked porcine dermal collagen. Cross-linking improves the tensile strength and long-term durabilitybut decreases pliability.

PriMatrix™ (TEI Biosciences) is a xenogeneic ADM processed from fetal bovine dermis. It was cleared for marketing by FDA through the 510(k) process for partial- and full-thickness wounds; diabetic, pressure, and venous stasis ulcers; surgical wounds; and tunneling, draining, and traumatic wounds. FDA product code: KGN.

SurgiMend® PRS (TEI Biosciences) is a xenogeneic ADM processed from fetal bovine dermis.

Strattice™ Reconstructive Tissue Matrix (LifeCell Corp.) is a xenogenic non-cross-linked porcine-derived ADM. There are pliable and firm versions, which are stored at room temperature and come fully hydrated.

Oasis™ Wound Matrix (Cook Biotech) is a collagen scaffold (extracellular matrix) derived from porcine small intestinal submucosa. In 2000, it was cleared for marketing by FDA through the 510(k) process for the management of partial- and full-thickness wounds, including pressure ulcers, venous ulcers, diabetic ulcers, chronic vascular ulcers, tunneled undermined wounds, surgical wounds, trauma wounds, and draining wounds. FDA Product code: KGN.

Living Cell Therapy

Apligraf® (Organogenesis) is a bilayered living cell therapy composed of an epidermal layer of living human keratinocytes and a dermal layer of living human fibroblasts. Apligraf® is supplied as needed, in 1 size, with a shelf-life of 10 days. In 1998, it was approved by FDA for use in conjunction with compression therapy for the treatment of noninfected, partial- and full-thickness skin ulcers due to venous insufficiency and in 2001 for full-thickness neuropathic diabetic lower-extremity ulcers nonresponsive to standard wound therapy. FDA product code: FTM.

Dermagraft® (Organogenesis) is composed of cryopreserved human-derived fibroblasts and collagen derived from newborn human foreskin and cultured on a bioabsorbable polyglactin mesh scaffold.Dermagraft has been approved by FDA for repair of diabetic foot ulcers. FDA product code: PFC.

TheraSkin® (Soluble Systems) is a cryopreserved split-thickness human skin allograft composed of living fibroblasts and keratinocytes and an extracellular matrix in epidermal and dermal layers. TheraSkin® is derived from human skin allograft supplied by tissue banks compliant with the American Association of Tissue Banks and FDA guidelines. It is considered a minimally processed human cell, tissue, and cellular- and tissue based product by FDA.

Epicel® (Genzyme Biosurgery) is an epithelial autograft composed of a patient’s own keratinocytes cultured ex vivo and is FDA-approved under a humanitarian device exemption for the treatment of deep dermal or full-thickness burns comprising a total body surface area of 30% or more. It may be used in conjunction with split-thickness autografts or alone in patients for whom split-thickness autografts may not be an option due to the severity and extent of their burns. FDA product code: OCE.

OrCel™ (Forticell Bioscience; formerly Composite Cultured Skin) is an absorbable allogeneic bilayered cellular matrix, made of bovine collagen, in which human dermal cells have been cultured. It was approved by FDA premarket approval for healing donor site wounds in burn victims and under a humanitarian device exemption (HDE) for use in patients with recessive dystrophic epidermolysis bullosa undergoing hand reconstruction surgery to close and heal wounds created by the surgery, including those at donor sites. FDA product code: ODS.

Biosynthetic Products

Biobrane®/Biobrane-L (Smith & Nephew) is a biosynthetic wound dressing constructed of a silicon film with a nylon fabric partially imbedded into the film. The fabric creates a complex 3-dimensional structure of trifilament thread, which chemically binds collagen. Blood/sera clot in the nylon matrix, adhering the dressing to the wound until epithelialization occurs. FDA product code: FRO.

Integra® Dermal Regeneration Template (also marketed as Omnigraft Dermal Regeneration Matrix; Integra LifeSciences) is a bovine, collagen/glycosaminoglycan dermal replacement covered by a silicone temporary epidermal substitute. It was approved by FDA for use in the postexcisional treatment of life-threatening full-thickness or deep partial-thickness thermal injury where sufficient autograft is not available at the time of excision or not desirable because of the physiologic condition of the patient and for certain diabetic foot ulcers. Integra® Matrix Wound Dressing and Integra® Meshed Bilayer Wound Matrix are substantially equivalent skin substitutes and were cleared for marketing by FDA through the 510(k) process for other indications. Integra® Bilayer Matrix Wound Dressing (Integra LifeSciences) is designed to be used in conjunction with negative pressure wound therapy. The meshed bilayer provides a flexible wound covering and allows drainage of wound exudate. FDA product code: MDD.

TransCyte™ (Advanced Tissue Sciences) consists of human dermal fibroblasts grown on nylon mesh, combined with a synthetic epidermal layer and was approved by FDA in 1997. TransCyte is intended as a temporary covering over burns until autografting is possible. It can also be used as a temporary covering for some burn wounds that heal without autografting.

Synthetic Products

Suprathel® (PolyMedics Innovations) is a synthetic copolymer membrane fabricated from a tripolymer of polylactide, trimethylene carbonate, and s-caprolactone. It is used to provide temporary coverage of superficial dermal burns and wounds. Suprathel® is covered with gauze and a dressing that is left in place until the wound has healed.

Related Policies

  • Recombinant and Autologous Platelet-Derived Growth Factors as a Treatment of Wound Healing and Other Non-Orthopedic Conditions (Policy #004 in the Treatment Section)
  • Amniotic Membrane and Amniotic Fluid (Policy #150 in the Surgery Section)

Policy:
(NOTE: For Medicare Advantage, Medicaid and FIDE-SNP, please refer to the Coverage Sections below for coverage guidance.)


1. Breast reconstructive surgery using allogeneic acellular dermal matrix products* (including each of the following: AlloDerm®, Cortiva® [AlloMax™], AlloMend®, DermACELL™, DermaMatrix™, FlexHD®, FlexHD® Pliable™, Graftjacket®; see Policy Guidelines) is considered medically necessary,
     when there is insufficient tissue expander or implant coverage by the pectoralis major muscle and additional coverage is required,
     when there is viable but compromised or thin postmastectomy skin flaps that are at risk of dehiscence or necrosis, or
     the inframammary fold and lateral mammary folds have been undermined during mastectomy and reestablishment of these landmarks is needed.

2. Treatment of chronic, noninfected, full-thickness diabetic lower-extremity ulcers using the following tissue-engineered skin substitutes is considered medically necessary.
     AlloPatch®*
     Apligraf®**
     Dermagraft®**
     Integra® Omnigraft Dermal Regeneration Matrix (also known as Omnigraft) and Integra Flowable Wound Matrix.

3. Treatment of chronic, noninfected, partial- or full-thickness lower-extremity skin ulcers due to venous insufficiency, which have not adequately responded following a 1-month period of conventional ulcer therapy, using the following tissue-engineered skin substitutes is considered medically necessary.
     Apligraf®**
     Oasis™ Wound Matrix***

4. Treatment of dystrophic epidermolysis bullosa using the following tissue-engineered skin substitutes is considered medically necessary.
     OrCel™ (for the treatment of mitten-hand deformity when standard wound therapy has failed and when provided in accordance with the Humanitarian Device Exemption (HDE) specifications of the U.S. Food and Drug Administration [FDA])****

5. Treatment of second- and third-degree burns using the following tissue-engineered skin substitutes is considered medically necessary.
     Epicel® (for the treatment of deep dermal or full-thickness burns comprising a total body surface area >30% when provided in accordance with the HDE specifications of the FDA)****
     Integra Dermal Regeneration Template™**

* Banked human tissue.
** FDA premarket approved.
*** FDA 510(k) clearance.
**** FDA-approved under an HDE.

6. All other uses of the bioengineered skin and soft tissue substitutes listed above are considered investigational.

7. All other skin and soft tissue substitutes not listed above are considered investigational, including, but not limited to:
     ACell® UBM Hydated/Lyophilized Wound Dressing
     AlloSkin™
     AlloSkin™ RT
     Aongen™ Collagen Matrix
     Architect® ECM, PX, FX
     ArthroFlex™ (FlexGraft)
     Atlas Wound Matrix
     Avagen Wound Dressing
     AxoGuard® Nerve Protector (AxoGen)
     Biobrane®/Biobrane-L
     CollaCare®
     CollaCare® Dental
     Collagen Wound Dressing (Oasis Research)
     CollaGUARD®
     CollaMend™
     CollaWound™
     Collexa®
     Collieva®
     Conexa™
     Coreleader Colla-Pad
     CorMatrix®
     Cymetra™ (Micronized AlloDerm™)
     Cytal™ (previously MatriStem®)
     Dermadapt™ Wound Dressing
     DermaPure™
     DermaSpan™
     DressSkin
     Durepair Regeneration Matrix®
     Endoform Dermal Template™
    ENDURAgen™
     Excellagen
     ExpressGraft™
     E-Z Derm™
     FlexiGraft®
     GammaGraft
     Graftjacket® Xpress, injectable
     Helicoll™
     Hyalomatrix®
     Hyalomatrix® PA
     hMatrix®
     Integra™ Bilayer Wound Matrix
     Keramatrix®
     Kerecis™
     MariGen™/Kerecis™ Omega3™
     MatriDerm®
     Matrix HD™
     Mediskin®
     MemoDerm™
     Microderm® biologic wound matrix
     NeoForm™
     NuCel
     Oasis® Burn Matrix
     Oasis® Ultra
     Pelvicol®/PelviSoft®
     Permacol™
     PriMatrix™
     Primatrix™ Dermal Repair Scaffold
     PuraPly™ Wound Matrix (previously FortaDerm™)
     PuraPly™ AM (Antimicrobial Wound Matrix)
     Puros® Dermis
     RegenePro™
     Repliform®
     Repriza™
     StrataGraft®
     Strattice™ (xenograft)
     Suprathel®
     SurgiMend®
     Talymed®
     TenoGlide™
     TenSIX™ Acellular Dermal Matrix
     TissueMend
     TheraForm™ Standard/Sheet
     TheraSkin®
     TruSkin™
     Veritas® Collagen Matrix
     XCM Biologic® Tissue Matrix
     XenMatrix™ AB.


Medicare Coverage:
Application of Bioengineered Skin Substitutes which are FDA-labeled or cleared for use in the types of wounds being treated are covered for Lower Extremity Chronic Non-Healing Wounds when LCD L35041 criteria is met. For additional information and eligibility, refer to Local Coverage Determination (LCD): Application of Bioengineered Skin Substitutes to Lower Extremity Chronic Non-Healing Wounds (L35041).Available at:https://www.cms.gov/medicare-coverage-database/details/lcd-details.aspx?LCDId=35041&ver=62&name=314*1&UpdatePeriod=765&bc=AAAAEAAAAAAA&.

Unless specifically FDA-labeled or cleared for use in the types of wounds being treated, the Bioengineered Skin Substitutes will be considered to be biologic dressings and part of the relevant Evaluation and Management (E/M) service provided and not separately reimbursed.

Skin substitute grafts will be allowed for the episode of wound care in compliance with FDA guidelines for the specific product not to exceed 10 applications or treatments. In situations where more than one specific product is used, it is expected that the number of applications or treatments will still not exceed 10

Per LCD L35041, the following are considered not reasonable and necessary and therefore will be denied:
    · Partial thickness loss with the retention of epithelial appendages is not a candidate for grafting or replacement, as epithelium will repopulate the deficit from the appendages, negating the benefit of overgrafting
    · Simultaneous use of more than one product for the episode of wound is not covered. Product change within the episode of wound is allowed, not to exceed the 10 application limit per wound per 12 week period of care.
    · Treatment of any chronic skin wound will typically last no more than twelve (12) weeks.
    · Repeat or alternative applications of skin substitute grafts are not considered medically reasonable and necessary when a previous full course of applications was unsuccessful. Unsuccessful treatment is defined as increase in size or depth of an ulcer or no change in baseline size or depth and no sign of improvement or indication that improvement is likely (such as granulation, epithelialization or progress towards closing) for a period of 4 weeks past start of therapy.
    · Retreatment of healed ulcers, those showing greater than 75% size reduction and smaller than .5 sq.cm, is not considered medically reasonable and necessary.
    · Skin substitute grafts are contraindicated and are not considered reasonable and necessary in patients with inadequate control of underlying conditions or exacerbating factors (e.g., uncontrolled diabetes, active infection, and active Charcot arthropathy of the ulcer extremity, vasculitis or continued tobacco smoking without physician attempt to effect smoking cessation).
    · Skin substitute grafts are contraindicated in patients with known hypersensitivity to any component of the specific skin substitute graft (e.g., allergy to avian, bovine, porcine, equine products).
    · Repeat use of surgical preparation services (CPT codes 15002, 15003, 15004, and 15005) in conjunction with skin substitute application codes will be considered not reasonable and necessary. It is expected that each wound will require the use of appropriate wound preparation code at least once at initiation of care prior to placement of the skin substitute graft.
    · Re-treatment within one (1) year of any given course of skin substitute treatment for a venous stasis ulcer or (diabetic) neuropathic foot ulcer is considered treatment failure and does not meet reasonable and necessary criteria for re-treatment of that ulcer with a skin substitute procedure.

Per LCD L35041, any HCPCS code that is not included in the LCD Group 2 list will not be separately reimbursed. HCPCS codes included in this list are inclusive of known FDA approved bioengineered wound dressings, skin substitutes, matrixes or scaffolding for chronic ulcer treatment as of publication. For additional information and eligibility, refer to Local Coverage Determination (LCD): Application of Bioengineered Skin Substitutes to Lower Extremity Chronic Non-Healing Wounds (L35041)

Medicaid Coverage:
For members enrolled in Medicaid and NJ FamilyCare plans, Horizon BCBSNJ applies the above medical policy.

FIDE-SNP Coverage:

For members enrolled in a Fully Integrated Dual Eligible Special Needs Plan (FIDE-SNP): (1) to the extent the service is covered under the Medicare portion of the member’s benefit package, the above Medicare Coverage statement applies; and (2) to the extent the service is not covered under the Medicare portion of the member’s benefit package, the above Medicaid Coverage statement applies.



Policy Guidelines: (Information to guide medical necessity determination based on the criteria contained within the policy statements above.)

Note that amniotic membrane and amniotic fluid products are addressed in a separate policy on 'Amniotic Membrane and Amniotic Fluid' (Policy #150 in the Surgery Section).

Clinical input indicated that the various acellular dermal matrix products used in breast reconstruction have similar efficacy. The products listed are those that have been identified for use in breast reconstruction. Additional acellular dermal matrix products may become available for this indication.


[RATIONALE: This policy was created in 2015 and has been updated regularly with searches of the MEDLINE database. The most recent literature update was performed through December 4, 2018.

The original review focused on the use of an allogeneic bioengineered skin substitutes in breast reconstructive surgery and was expanded in 2011 to address additional indications.

Evidence reviews assess the clinical evidence to determine whether the use of a technology improves the net health outcome. Broadly defined, health outcomes are length of life, quality of life, and ability to function¾including benefits and harms. Every clinical condition has specific outcomes that are important to patients and to managing the course of that condition. Validated outcome measures are necessary to ascertain whether a condition improves or worsens; and whether the magnitude of that change is clinically significant. The net health outcome is a balance of benefits and harms.

To assess whether the evidence is sufficient to draw conclusions about the net health outcome of a technology, 2 domains are examined: the relevance and the quality and credibility. To be relevant, studies must represent one or more intended clinical use of the technology in the intended population and compare an effective and appropriate alternative at a comparable intensity. For some conditions, the alternative will be supportive care or surveillance. The quality and credibility of the evidence depend on study design and conduct, minimizing bias and confounding that can generate incorrect findings. The randomized controlled trial (RCT) is preferred to assess efficacy; however, in some circumstances, nonrandomized studies may be adequate. RCTs are rarely large enough or long enough to capture less common adverse events and long-term effects. Other types of studies can be used for these purposes and to assess generalizability to broader clinical populations and settings of clinical practice.

The primary end points of interest for trials of wound closure are as follows, consistent with guidance from the U.S. Food and Drug Administration for industry in developing products for treatment of chronic cutaneous ulcer and burn wounds:

1.     Incidence of complete wound closure.

2.     Time to complete wound closure (reflecting accelerated wound closure).

3.     Incidence of complete wound closure following surgical wound closure.

4.     Pain control.

The following is a summary of key literature to date.

Breast Reconstruction

A variety of breast reconstruction techniques are used postmastectomy, including implant-based (immediate or delayed following use of a tissue expander) and those using autologous tissue flaps. Some of these techniques have been used with acellular dermal matrix (ADM) to provide additional support or tissue coverage. The literature on ADM for breast reconstruction consists primarily of retrospective, uncontrolled series and systematic reviews of these studies.

A 2013 study used data from the American College of Surgeon’s National Surgical Quality Improvement Program to compare ADM-assisted tissue expander breast reconstruction (n=1717) to submuscular tissue expander breast reconstruction (n=7442) after mastectomy.1 Complication rates did not differ significantly between the ADM-assisted (5.5%) and the submuscular tissue expander groups (5.3%; p=0.68). Rates of reconstruction-related complications, major com­plications, and 30-day reoperation did not differ significantly between cohorts.

Systematic Reviews

A 2016 meta-analysis by Lee and Mun included 23 studies (total N=6199 cases) on implant-based breast reconstruction that were published between February 2011 and December 2014.2, The analysis included an RCT and 3 prospective comparative cohort studies; the remainder was retrospective comparative cohort studies. Use of ADM did not affect the total complication rate (see Table 1). ADM significantly increased the risk of major infection, seroma, and flap necrosis, but reduced risks of capsular contracture and implant malposition. Use of ADM allowed for significantly greater intraoperative expansion (mean difference [MD], 79.63; 95% confidence interval [CI], 41.99 to 117.26; p<0.001) and percentage of intraoperative filling (MD=13.30; 95% CI, 9.95 to 16.65; p<0.001), and reduced the frequency of injections to complete expansion (MD = -1.56; 95% CI, -2.77 to -0.35; p=0.01).

Table 1. Meta-Analysis of Breast Reconstruction Outcomes With and Without ADM
Outcome MeasureRelative Risk95% Confidence Intervalp
Infection1.421.02 to 1.990.04
Seroma1.411.12 to 1.780.004
Mastectomy flap necrosis1.441.11 to 1.870.006
Unplanned return to the operating room1.090.63 to 1.90NS
Implant loss1.000.68 to 1.48NS
Total complications1.080.87 to 1.34NS
Capsular contracture0.260.15 to 0.47<0.001
Implant malposition0.210.07 to 0.590.003
Adapted from Lee and Mun (2016).2,
ADM: acellular dermal matrix; NS: not significant.

AlloDerm

Randomized Controlled Trials

In 2012, McCarthy et al reported on a multicenter, blinded RCT of AlloDerm in 2-stage expander/implant reconstruction.3, Seventy patients were randomized to AlloDerm ADM-assisted tissue expander/implant reconstruction or to submuscular tissue expander/implant placement. The trial was adequately powered to detect clinically significant differences in immediate postoperative pain but underpowered to detect the secondary end point of pain during tissue expansion. There were no significant differences between the groups in the primary outcomes of immediate postoperative pain (54.6 AlloDerm vs 42.8 controls on a 100-point visual analog scale) or pain during the expansion phase (17.0 AlloDerm vs 4.6 controls), or in the secondary outcome of rate of tissue expansion (91 days AlloDerm vs 108 days controls) and patient-reported physical well-being. There was no significant difference in adverse events, although the total number of adverse events was small.

Comparisons Between Products

AlloDerm vs AlloMax

Hinchcliff et al (2017) conducted an RCT that compared AlloDerm with AlloMax (n=15 each) for implant-based breast reconstruction.4, Complications were assessed 7, 14, and 30 days postoperatively and biopsies of the ADMs were taken during implant exchange. Vessel density in the AlloMax biopsies was higher than in the AlloDerm biopsies. Complications were reported in 26.1% of AlloMax cases and 8.0% of AlloDerm cases; these complication rates did not differ statistically with the 30 patients in this trial.

AlloDerm vs DermaMatrix

Mendenhall et al (2017) conducted an RCT that compared AlloDerm with DermaMatrix in 111 patients (173 breasts).5, There were no significant differences in overall rates of complications (AlloDerm, 15.4%; DermaMatrix, 18.3%; p=0.8) or implant loss (AlloDerm, 2.2%; DermaMatrix, 3.7%; p=0.5) between the 2 ADMs.

AlloDerm vs FlexHD

A 2014 retrospective review by Liu et al compared complication rates following breast reconstruction with AlloDerm or FlexHD in 382 consecutive women (547 breasts).6, Eighty-one percent of the sample was immediate reconstruction: 165 used AlloDerm and 97 used FlexHD. Mean follow-up was 6.4 months. Compared with breast reconstruction without the use of AlloDerm or FlexHD, ADM had a higher rate of delayed healing (20.2% vs 10.3%), although this finding might have been related to differences in fill volumes. In univariate analysis, there were no significant differences in complications (return to operating room, surgical site infection, seroma, hematoma, delayed healing, implant loss) between AlloDerm and FlexHD. In multivariate analysis, there were no significant differences between AlloDerm and FlexHD for the return to the operating room, surgical site infection, seroma, or delayed healing. Independent risk factors for implant loss included the use of FlexHD, single-stage reconstruction, and smoking.

AlloDerm vs FlexHD Pliable and DermACELL

Chang and Liu (2017) reported on a prospective comparison of FlexHD Pliable (32 breasts), AlloDerm (22 breasts), and DermACELL (20 breasts) in breast reconstruction.7, The choice of ADM was based on different years when each ADM was available for use at the investigators’ institution; patient demographics were comparable between groups. The pieces of ADM used were all the same size (8 × 16 cm) to eliminate an effect of size on outcomes. The time to drain removal was longer with AlloDerm (26 days) than with FlexHD (20 days) or DermACELL (15 days; p=0.001). Complications were low (4 in the Flex Pliable group, 2 in the AlloDerm group, 1 in the DermACELL group), with no significant differences between groups. At the time of exchange for a permanent implant or free flap reconstruction, all grafts had completely incorporated into the mastectomy skin flaps. No patients developed complications requiring removal of the ADM.

Pittman et al (2017) reported a retrospective pilot study of the use of AlloDerm (50 breasts) and DermACELL (50 breasts).8, The choice of ADM was based on products available during different years and patient demographics were similar between the 2 groups. Patients in the DermACELL group had a significantly lower incidence of “red breast syndrome” (0% vs 26%, p=0.001) and fewer days until drain removal (15.8 days vs 20.6 days, p=0.017). There were no significant differences in the rates of other complications.

Strattice

Dikmans et al (2017) reported on early safety outcomes from an open-label multicenter RCT that compared porcine ADM-assisted 1-stage expansion with 2-stage implant-based breast reconstruction (see Table 2).9 One-stage breast reconstruction with porcine ADM was associated with a higher risk of surgical complications, reoperation, and with removal of implant, ADM, or both (see Table 3). The trial was stopped early due to safety concerns, but it cannot be determined from this study design whether the increase in complications was due to the use of the xenogenic ADM or to the comparison between 1-stage and 2-stage reconstruction.

Table 2. Summary of Key RCT Characteristics
    Interventions
AuthorCountriesSitesDatesParticipantsActiveComparator
Dikmans et al (2017)9,EU82013-2015Women intending to undergo skin-sparing mastectomy and immediate IBBR59 patients (91 breasts) undergoing 1-stage IBBR with ADM62 women (92 breasts) undergoing 2-stage IBBR
ADM: acellular dermal matrix; IBBR: implant-based breast reconstruction; RCT: randomized controlled trial.

Table 3. Summary of Key RCT Outcomes
StudySurgical ComplicationsSevere Adverse EventsReoperationRemoval of Implant, ADM, or Both
Dikmans et al (2017)9,    
1-stage with ADM, n (%)27 (46)26 (29)22 (37)24 (26)
2-stage with ADM, n (%)11 (18)5 (5)9 (15)4 (5)
OR (95% CI)3.81 (2.67 to 5.43) 3.38 (2.10 to 5.45)8.80 (8.24 to 9.40)
p<0.001 <0.001<0.001
ADM: acellular dermal matrix; CI: confidence interval; OR: odds ratio; RCT: randomized controlled trial.

Section Summary: Breast Reconstruction

Results of a systematic review found no difference in overall complication rates between ADM allograft and standard procedures for breast reconstruction. Although reconstructions with ADM have been reported to have higher seroma, infection, and necrosis rates than reconstructions without ADM, rates of capsular contracture and malposition of implants may be reduced. Thus, in cases where there is limited tissue coverage, the available studies may be considered sufficient to permit informed decision-making about risks and benefits of using allogeneic ADM for breast reconstruction.

Tendon Repair

Graftjacket

In 2012, Barber et al reported an industry-sponsored multicenter RCT of augmentation with Graftjacket human ADM for arthroscopic repair of large (>3 cm) rotator cuff tears involving 2 tendons.10, Twenty-two patients were randomized to Graftjacket augmentation and 20 patients to no augmentation. At a mean follow-up of 24 months (range, 12-38 months), the American Shoulder and Elbow Surgeons score improved from 48.5 to 98.9 in the Graftjacket group and from 46.0 to 94.8 in the control group (p=0.035). The Constant score improved from 41 to 91.9 in the Graftjacket group and from 45.8 to 85.3 in the control group (p=0.008). The University of California, Los Angeles score did not differ significantly between groups. Gadolinium-enhanced magnetic resonance imaging (MRI) scans showed intact cuffs in 85% of repairs in the Graftjacket group and 40% of repairs in the control group. However, no correlation was found between MRI findings and clinical outcomes. Rotator cuff retears occurred in 3 (14%) patients in the Graftjacket group and 9 (45%) patients in the control group.

Section Summary: Tendon Repair

One small RCT was identified that found improved outcomes with Graftjacket ADM allograft for rotator cuff repair. Although results of this trial were promising, additional study with a larger number of patients is needed to evaluate consistency of findings and determine the effects of this technology with greater certainty.

Surgical Repair of Hernias or Parastomal Reinforcement

A 2013 systematic review evaluated the clinical effectiveness of acellular collagen-based scaffolds for the repair of incisional hernias.11, The bioprosthetic materials could be harvested from bovine pericardium, human cadaveric dermis, porcine small intestine mucosa, porcine dermal collagen, or bovine dermal collagen. Products included in the search were Surgisis, Tutomesh, Veritas, AlloDerm, FlexHD, AlloMax, CollaMend, Permacol, Strattice, FortaGen, ACell, DermaMatrix, XenMatrix, and SurgiMend. Sixty publications with 1212 repairs were identified and included in the review, although meta-analysis could not be performed. There were 4 level III studies (2 AlloDerm, 2 Permacol); the remainder was level IV or V. The largest number of publications were on AlloDerm (n=27) and Permacol (n=18). No publications on incisional hernia repair were identified for AlloMax, FortaGen, DermaMatrix, or ACell. The overall incidence of a surgical site occurrence (eg, postoperative infection, seroma/hematoma, pain, bulging, dehiscence, fistula, mechanical failure) was 82.6% for porcine small intestine mucosa, 50.7% for xenogenic dermis, 48.3% for human dermis, and 6.3% for xenogenic pericardium. No comparative data were identified that could establish superiority to permanent synthetic meshes.

AlloDerm as an Overlay

In 2007, Espinosa-de-los-Monteros et al retrospectively reviewed 39 abdominal wall reconstructions with AlloDerm performed in 37 patients and compared them with 39 randomly selected cases.12, They reported a significant decrease in recurrence rates when human cadaveric acellular dermis was added as an overlay to primary closure plus rectus muscle advancement and imbrication in patients with medium-sized hernias. However, no differences were observed when adding human cadaveric acellular dermis as an overlay to patients with large-size hernias treated with underlay mesh.

Comparisons Between Products

AlloDerm vs Surgisis Gold

Gupta et al (2006) compared the efficacy and complications associated with use of AlloDerm and Surgisis bioactive mesh in 74 patients who underwent ventral hernia repair.13, The first 41 procedures were performed using Surgisis Gold 8-ply mesh formed from porcine small intestine submucosa, and the remaining 33 patients had ventral hernia repair with AlloDerm. Patients were seen 7 to 10 days after discharge from the hospital and at 6 weeks. Any signs of wound infection, diastasis, hernia recurrence, changes in bowel habits, and seroma formation were evaluated. The use of the AlloDerm mesh resulted in 8 (24%) hernia recurrences. Fifteen (45%) of the AlloDerm patients developed a diastasis or bulging at the repair site. Seroma formation was only a problem in 2 patients.

AlloDerm vs FlexHD

A 2013 study compared AlloDerm with FlexHD for complicated hernia surgery.14, From 2005 to 2007, AlloDerm was used to repair large (>200 cm2) symptomatic complicated ventral hernias that resulted from trauma or emergency surgery (n=55). From 2008 to 2010, FlexHD was used to repair large, complicated ventral hernias in patients meeting the same criteria (n=40). The 2 groups were comparable at baseline. At 1 year follow-up, all AlloDerm patients were diagnosed with hernia recurrence (abdominal laxity, functional recurrence, true recurrence) requiring a second repair. Eleven (31%) patients in the FlexHD group required a second repair. This comparative study is limited by the use of nonconcurrent comparisons, which is prone to selection bias and does not control for temporal trends in outcomes.

FlexHD vs Strattice

Roth et al (2017) reported on a prospective study assessing clinical and quality of life outcomes following complex hernia repair with a human (FlexHD) or porcine (Strattice) ADM.15, The study was funded by the Musculoskeletal Transplant Foundation, which prepares and supplies FlexHD. Patients were enrolled if they had a hernia at least 6 cm in the transverse dimension, active or prior infection of the abdominal wall, and/orenterocutaneous fistula requiring mesh removal. Eighteen (51%) of the 35 patients had undergone a previous hernia repair. After abdominal wall repair with the ADM, 20 (57%) patients had a surgical site occurrence, and nearly one-third had hospital readmission. The type of biologic material did not impact hernia outcomes. There was no comparison with synthetic mesh in this study, limiting interpretation.

Strattice vs Synthetic Mesh

In 2014, Bellows et al reported early results of an industry-sponsored multicenter RCT that compared Strattice (non-cross-linked porcine ADM, n=84) with a standard synthetic mesh (n=88) for the repair of inguinal hernias.16, The trial was designed by the surgeons and was patient- and assessor-blinded to reduce risk of bias. Blinding continued through 2 years of follow-up. The primary outcome was resumption of activities of daily living at 1 year. Secondary outcomes included complications, recurrences, or chronic pain (ie, pain that did not disappear by 3 months postsurgery). At 3-month follow-up, there were no significant differences in either the occurrence or type of wound events (relative risk, 0.98; 95% CI, 0.52 to 1.86). Painwas reduced from 1 to 3 days postoperative in the group treated with Strattice, but at 3 month follow-up pain scores did not differ significantly between groups.

Strattice vs No Reinforcement

Also in 2014, the PRISM Study Group reported a multicenter, double-blinded, randomized trial of Strattice for parastomal reinforcement in patients undergoing surgery for permanent abdominal wall ostomies.17, Patients were randomized to standard stoma construction with no reinforcement (n=58) or stoma construction with Strattice as parastomal reinforcement (n=55). At 24-month follow-up (n=75), the incidence of parastomal hernias was similar for the 2 groups (13.2% of controls, 12.2% of study group).

Section Summary: Surgical Repair of Hernias or Parastomal Reinforcement

Current evidence does not support a benefit of ADMs in hernia repair or prevention of parastomal hernia. Additional RCTs are needed to compare biologic mesh with synthetic mesh and to determine if there is a patient population that would benefit from these products.

Diabetic Lower-Extremity Ulcers

Systematic Reviews

A 2016 Cochrane review evaluated skin substitutes for the treatment of diabetic foot ulcers.18, Seventeen trials (total N=1655 participants) were included in the meta-analysis. Most trials identified were industry-sponsored, and an asymmetric funnel plot indicated publication bias. Pooled results of published trials found that skin substitutes increased the likelihood of achieving complete ulcer closure compared with standard of care (SOC) alone (relative risk, 1.55; 95% CI, 1.30 to 1.85). Use of skin substitutes also led to a statistically significant reduction in amputations (relative risk, 0.43; 95% CI, 0.23 to 0.81), although the absolute risk difference was small. Analysis by individual products found a statistically significant benefit on ulcer closure for Apligraf, EpiFix, and Hyalograft-3D. The products that did not show a statistically significant benefit for ulcer closure were Dermagraft, Graftjacket, Kaloderm, and OrCel. Individual RCTs are described next.

Martinson and Martinson (2016) conducted an industry-sponsored analysis of Medicare claims data (13,193 treatment episodes) to compare efficacy and cost of skin substitutes for the management of diabetic foot ulcers.19, Included in the analysis were treatment episodes with Apligraf (37%), Dermagraft (42%), Oasis (19%), and Cytal (MatriStem, 2%). The mean number of applications was 3.24 for Apligraf, 4.48 for Oasis, 5.53 for Cytal, and 5.96 for Dermagraft. All comparisons were statistically significant. Healing at 90 days was modestly but statistically higher for Oasis (63%) and Cytal (62%) than for Apligraf (58%) or Dermagraft (58%). Amputation rates were similar after treatment with the 4 products, ranging from 1.3% for Oasis to 2.1% for Cytal.

Guo et al (2017) reported a systematic review of ADM for the treatment diabetic foot ulcer.20,Most data were from an RCT of Integra Dermal Regeneration Template, which is a bilayer product with the outer layer composed of a thin silicone film and not a pure ADM.

Apligraf, Dermagraft, AlloPatch, Integra Dermal Regeneration Template, or Integra Flowable Wound Matrix

Apligraf

Veves et al (2001) reported on a randomized prospective trial on the effectiveness of Apligraf (previously called Graftskin), a living skin equivalent, in treating noninfected nonischemic chronic plantar diabetic foot ulcers.21, The trial involved 24 centers in the United States; 208 patients were randomized to ulcer treatment with Apligraf (112 patients) or saline-moistened gauze (96 patients, control group). Standard state-of-the-art adjunctive therapy, including extensive surgical débridement and adequate foot off-loading, was provided in both groups. Apligraf was applied at the beginning of the study and weekly thereafter for a maximum of 4 weeks (maximum of 5 applications) or earlier if complete healing occurred. At the 12-week follow-up visit, 63 (56%) Apligraf-treated patients achieved complete wound healing compared with 36 (38%) in the control group (p=0.004). The Kaplan-Meier method median time to complete closure was 65 days for Apligraf, significantly lower than the 90 days observed in the control group (p=0.003). The rates of adverse reactions were similar between groups, except osteomyelitis and lower-limb amputations, both of which were less frequent in the Apligraf group. Trialists concluded that application of Apligraf for a maximum of 4 weeks resulted in higher healing rates than state-of-the-art treatment and was not associated with any significant adverse events. This trial was reviewed in a 2001 TEC Assessment, which concluded that Apligraf, in conjunction with good local wound care, met the TEC criteria for the treatment of diabetic ulcers that fail to respond to conservative management.22,

Steinberg et al (2010) reported on a study of 72 subjects from Europe and Australia that assessed the safety and efficacy of Apligraf in the treatment of noninfected diabetic foot ulcers.23, Study design and patient population were similar to the 208-subject U.S. study (previously described), which led to Food and Drug Administration (FDA) approval of Apligraf for the treatment of diabetic foot ulcers. For these studies, subjects with noninfected neuropathic diabetic foot ulcers present for at least 2 weeks were enrolled in prospective, multicenter, open-label RCTs that compared Apligraf use plus standard therapy (sharp débridement, standard wound care, off-loading) with standard therapy alone. Pooling of data was performed because of the similarity and consistency of the 2 studies. Efficacy and safety results were consistent across studies independent of mean ulcer duration, which was significantly longer in the European study (21 months vs10 months in the U.S. study). Reported adverse events by 12 weeks were comparable across treatment groups in the 2 studies. Efficacy measures demonstrated superiority of Apligraf treatment over control-treated groups in both studies. Combining the data from both studies, 55.2% (80/145) of Apligraf subjects had complete wound closure by 12 weeks, compared with 34.3% (46/134) of control subjects (p<0.001), and Apligraf subjects had a significantly shorter time to complete wound closure (p<0.001). The authors concluded that both the EU and U.S. studies exhibited superior efficacy and comparable safety for subjects treated with Apligraf compared with control subjects and that the studies provided evidence of the benefit of Apligraf in treating diabetic foot ulcer.

In 2010, Kirsner et al analyzed 2517 patients with diabetic neuropathic foot ulcers treated between 2001 and 2004.24, This retrospective analysis used a wound care database; the patients received advanced biologic therapy, specifically, Apligraf (446 patients), Regranex, or Procuren. The analysis found that advanced biologic therapy was used, on average, within 28 days from the first wound clinic visit and was associated with a median time to healing of 100 days. Wounds treated with engineered skin (Apligraf) as the first advanced biologic therapy were 31% more likely to heal than wounds first treated with topical recombinant growth factor (p<0.001) and 40% more likely to heal than those first treated with platelet releasate (p=0.01). Wound size, wound grade, duration of wound, and time to initiation of advanced biologic therapy affected the time to healing.

Dermagraft

A 2003 pivotal multicenter FDA-regulated trial randomized 314 patients with chronic diabetic ulcers to Dermagraft (human-derived fibroblasts cultured on mesh) or control.25, Over the 12-week study, patients received up to 8 applications of Dermagraft. All patients received pressure-reducing footwear and were encouraged to stay off their study foot as much as possible. At 12 weeks, the median percent wound closure for the Dermagraft group was 91% compared with 78% for the control group. Ulcers treated with Dermagraft closed significantly faster than ulcers treated with conventional therapy. No serious adverse events were attributed to Dermagraft. Ulcer infections developed in 10.4% of the Dermagraft patients compared with 17.9% of the control patients. Together, there was a lower rate of infection, cellulitis, and osteomyelitis in the Dermagraft-treated group (19% vs 32.5%). A 2015 retrospective analysis of the trial data found a significant reduction in amputation/bone resection rates with Dermagraft (5.5% vs 12.6%, p=0.031).26, Of the 28 cases of amputation/bone resection, 27 were preceded by ulcer-related infection.

AlloPatch

AlloPatch Pliable human reticular acellular dermis was compared with SOC in an industry-sponsored multicenter trial by Zelen et al (2017, 2018).27,28, The initial trial with 20 patients per group was extended to determine the percent healing at 6 weeks with 40 patients per group. Healing was evaluated by the site investigator and confirmed by an independent panel. At 6 weeks, 68% (27/40) of wounds treated using AlloPatch had healed compared with 15% (6/40) in the SOC-alone group (p<0.001). At 12 weeks, 80% (32/40) of patients in the AlloPatch group had healed compared to 30% (12/40) in the control group. Mean time to heal within 12 weeks was 38 days (95% CI: 29-47 days) for the HR-ADM group and 72 days (95% CI: 66-78 days) for the SOC group (p < 0.001).

Integra Omnigraft Dermal Regeneration Template or Integra Flowable Wound Matrix

Integra Dermal Regeneration Template is a biosynthetic skin substitute that is FDA-approved for life-threatening thermal injury. The FOUNDER (Foot Ulcer New Dermal Replacement) multicenter study (32 sites) assessed Integra Dermal Regeneration Template (marketed as Omnigraft) for chronic nonhealing diabetic foot ulcers under an FDA-regulated investigational device exemption.29, A total of 307 patients with at least 1 chronic diabetic foot ulcer were randomized to treatment with the Integra Template or a control condition (sodium chloride gel 0.9%). Treatment was given for 16 weeks or until wound closure. There was a modest increase in wound closure with the Integra Template (51% vs 32%, p=0.001) and a shorter median time to closure (43 days vs 78 days, p=0.001). There was a strong correlation between investigator-assessed and computerized planimetry assessment of wound healing (r=0.97). Kaplan-Meier analysis showed the greatest difference between groups in wound closure up to 10 weeks, with diminishing differences after10 weeks. Trial strengths included adequate power to detect an increase in wound healing of 18%, which was considered to be clinically significant, secondary outcomes of wound closure and time to wound closure by computerized planimetry, and ITT analysis.

Integra Flowable Wound Matrix is composed of a porous matrix of cross-linked bovine tendon collagen and glycosaminoglycan. It is supplied as a granular product that is mixed with saline. Campitiello et al (2017) published an RCT that compared the flowable matrix with wet dressing in 46 patients who had Wagner grade 3 diabetic foot ulcers.30, The ulcers had developed over 39 weeks. Complete healing at 6 weeks was achieved in significantly more patients in the Integra Flowable Wound Matrix group than in the control group, while the risk of rehospitalization and major amputation was reduced with Integra Flowable Wound Matrix (see Table 4).

Table 4. Probability of Wound Healing With IFWM vs SOC
StudyComplete Wound HealingRehospitalizationMajor Amputation
Campitiello et al (2017)30,   
IFWM, n (%)20 (86.95)2 (6.69)1 (4.34)
SOC, n (%)12 (52.17)10 (43.47)7 (30.43)
RR (95% CI)1.67 (1.09 to 2.54)0.10 (0.01 to 0.72)0.16 (0.02 to 1.17)
p0.0100.0010.028
CI: confidence interval; IFWM: Integra Flowable Wound Matrix; RR: relative risk; SOC: standard of care.

 Section Summary: Apligraf, Dermagraft, AlloPatch, or Integra for Diabetic Lower-Extremity Ulcers

RCTs have demonstrated the efficacy of Apligraf, Dermagraft, AlloPatch, Integra Dermal Regeneration Template, and Integra Flowable Wound Matrix over SOC for the treatment of diabetic lower-extremity ulcers.

Bioengineered Skin Substitutes Other Than Apligraf, Dermagraft, AlloPatch, or Integra

Graftjacket Regenerative Tissue Matrix

Brigido et al (2004) reported a small (N=40) randomized pilot study comparing Graftjacket with conventional treatment for chronic nonhealing diabetic foot ulcers.31, Control patients received conventional therapy with débridement, wound gel with gauze dressing, and off-loading. Graftjacket patients received surgical application of the scaffold using skin staples or sutures and moistened compressive dressing. A second graft application was necessary after the initial application for all patients in the Graftjacket group. Preliminary one month results showed that, after a single treatment, ulcers treated with Graftjacket healed at a faster rate than conventional treatment. There were significantly greater decreases in wound length (51% vs 15%), width (50% vs 23%), area (73% vs 34%), and depth (89% vs 25%), respectively. With follow-up to four weeks, no data were reported on the proportion with complete closure or the mean time to heal. All grafts were incorporated into the host tissue.

Reyzelman et al (2009) reported an industry-sponsored multicenter randomized study that compared a single application of Graftjacket with SOC in 86 patients with diabetic foot ulcers.32, Eight patients, six in the study group and two in the control group, did not complete the trial. At 12 weeks, complete healing was observed in 69.6% of the Graftjacket group and 46.2% of controls. After adjusting for ulcer size at presentation, a statistically significant difference in nonhealing rate was calculated, with odds of healing 2.0 times higher in the study group. Mean healing time was 5.7 weeks for the Graftjacket group vs 6.8 weeks for the control group. The authors did not report whether this difference was statistically significant. Median time to healing was 4.5 weeks for Graftjacket (range, 1-12 weeks) and 7.0 weeks for control (range, 2-12 weeks). Kaplan-Meier method survivorship analysis for time to complete healing at 12 weeks showed a significantly lower nonhealing rate for the study group (30.4%) than for the control group (53.9%). The authors commented that a single application of Graftjacket, as used in this study, was often sufficient for complete healing. Conclusions drawn from this study are limited by the small study population and differences in ulcer size at baseline. Questions also remain whether the difference in mean time to healing is statistically or clinically significant.

In 2015, Reyzelman and Bazarov33, reported an industry-sponsored meta-analysis of Graftjacket for diabetic foot ulcers that included the 2 studies described above and a third RCT by Brigido (2006)34, with 28 patients (total N=154 patients). The time to heal was estimated for the 2004 Brigido study, based on the average wound reduction per week. The estimated difference in time to heal was considerably larger for Brigido’s 2004 study (-4.30 weeks) than for the other 2 studies that measured the difference in time to heal (-1.58 weeks and -1.10 weeks). Analysis of the proportion of wounds that healed included Brigido (2006) and Reyzelman et al (2009). The odds ratio in the smaller study by Brigido was considerably larger, with a lack of precision in the estimate (odds ratio, 15.0; 95% CI, 2.26 to 99.64), and the combined odds (3.75; 95% CI, 1.72 to 8.19) was not significant when analyzed using a random-effects model. Potential sources of bias, noted by Reyzelman and Bazarov, included publication and reporting biases, study selection biases, incomplete data selection, post hoc manipulation of data, and subjective choice of analytic methods. Overall, results of these studies do not provide convincing evidence that Graftjacket is more effective than SOC for healing diabetic foot ulcers.

DermACELL vs Graftjacket Regenerative Tissue Matrix or SOC

DermACELL and Graftjacket are both composed of human ADM. In 2016, Walters et al reported on a multicenter randomized comparison of DermACELL, Graftjacket, or SOC (2:1:2 ratio) in 168 patients with diabetic foot ulcers.35, The study was sponsored by LifeNet Health, a nonprofit organ procurement association and processor for DermACELL. At 16 weeks, the proportion of completely healed ulcers was 67.9% for DermACELL, 47.8% for Graftjacket, and 48.1% for SOC. The 20% difference in completely healed ulcers was statistically significant for DermACELL vs SOC (p=0.039). The mean time to complete wound closure did not differ significantly for DermACELL (8.6 weeks), Graftjacket (8.6 weeks), and SOC (8.7 weeks).

A second report from this study was published in 2017.36, This analysis compared DermACELL with SOC and did not include the Graftjacket arm. The authors reported that either 1 or 2 applications DermACELL led to a greater proportion of wounds healed compared with SOC in per protocol analysis (see Table 5), but there was no significant difference between DermACELL (1 or 2 applications) and SOC when analyzed by intention-to-treat. For the group of patients who received only a single application, the percentage of patients who achieved complete wound healing was significantly higher than SOC at 16 and 24 weeks, but not at 12 weeks. Although reported as ITT analysis, results were analyzed only for the group who received a single application of DermACELL. This would not typically be considered ITT unless the number of DermACELL applications was prespecified.

Table 5. Probability of Wound Healing in Per Protocol Analysis of DermACELL vs SOC
StudySingle Application1 or 2 Applications
 % With Wound Healing at 12 Wk% With Wound Healing at 16 Wk% With Wound Healing at 24 Wk% With Wound Healing at 12 Wk% With Wound Healing at 16 Wk% With Wound Healing at 24 Wk
Cazzell et al (2017)36,     
DermACELL, %65.0%82.5%89.7%NR67.9%83.7%
SOC, %41.1%48.1%67.3%NR48.1%67.3%
HR (95% CI)1.97 
(1.1 to 3.5)
2.40 
(1.4 to 4.1)
2.11 
(1.3 to 3.5)
 1.72 
(1.04 to 2.83)
1.55 
(0.98 to 2.44)
p0.012<0.001<0.001NS0.0280.049
CI: confidence interval; HR: hazard ratio; NR; not reported; NS: not significant; SOC: standard of care.

TheraSkin vs Dermagraft

Sanders et al (2014) reported on a small (N=23) industry-funded randomized comparison of TheraSkin (cryopreserved human skin allograft with living fibroblasts and keratinocytes) and Dermagraft for diabetic foot ulcers.37 Wound size at baseline ranged from 0.5 to 18.02 cm2; the average wound size was about 5 cm2 and was similar for the 2 groups (p=0.51). Grafts were applied according to manufacturers’ instructions over the first 12 weeks of the study until healing, with an average of 4.4 TheraSkin grafts (every 2 weeks) compared with 8.9 Dermagraft applications (every week). At week 12, complete wound healing was observed in 63.6% of ulcers treated with TheraSkin and 33.3% of ulcers treated with Dermagraft (p<0.049). At 20 weeks, complete wound healing was observed in 90.9% of the TheraSkin-treated ulcers compared with 66.7% of the Dermagraft group (p=0.428).

TheraSkin vs Apligraf

DiDomenico et al (2011) compared TheraSkin with Apligraf for the treatment of diabetic foot ulcers in a small (N=29) RCT.38, The risk of bias in this study is uncertain, because reporting did not include a description of power analysis, statistical analysis, method of randomization, or blinding. The percentage of wounds closed at 12 weeks was 41.3% in the Apligraf group and 66.7% in the TheraSkin group. Results at 20 weeks were not substantially changed from those at 12 weeks, with 47.1% of wounds closed in the Apligraf group and 66.7% closed in the TheraSkin group. The percentage healed in the Apligraf group was lower than expected based on prior studies. The average number of grafts applied was similar for both groups (1.53 for Apligraf, 1.38 for TheraSkin). The low number of dressing changes may have influenced results, with little change in the percentage of wounds closed between 12 and 20 weeks. An adequately powered trial with blinded evaluation of wound healing and a standard treatment regimen would permit greater certainty on the efficacy of this product.

Cytal (MatriStem) vs Dermagraft

Frykberg et al (2017) reported a prespecified interim analysis of an industry-funded multicenter noninferiority trial of Cytal (a porcine urinary bladder-derived extracellular matrix) vs Dermagraft in 56 patients with diabetic foot ulcers.39, The mean duration of ulcers before treatment was 263 days (range, 30-1095 days). The primary outcome was the percent wound closure with up to 8 weeks of treatment using blinded evaluation of photographs. Intention-to-treat (ITT) analysis found complete wound closure in 5 (18.5%) wounds treated with Cytal compared with 2 (6.9%) wounds treated with Dermagraft (p=NS). Quality of life, measured by the Diabetic Foot Ulcer Scale, improved from 181.56 to 151.11 in the Cytal group and from 184.46 to 195.73 in the Dermagraft group (p=0.074). It should be noted that this scale is a subjective measure and patients were not blinded to treatment. Power analysis indicated that 92 patients would be required; further recruitment is ongoing for completion of the study.

PriMatrix

Kavros et al (2014) reported a prospective multicenter study of PriMatrix (a xenograft fetal bovine dermal collagen matrix) for the treatment of chronic diabetic foot ulcers in 55 patients.40, Average duration of ulcers before treatment was 286 days, and average wound area was 4.34 cm2. Of the 46 patients who completed the study, 76% healed by 12 weeks with an average of 2 applications of PriMatrix. For the ITT population, 64% of wounds healed by 12 weeks.

Karr (2011) published a retrospective comparison of PriMatrix and Apligraf in 40 diabetic foot ulcers.41, The first 20 diabetic foot ulcers matching the inclusion and exclusion criteria for each graft were compared. The criteria were: diabetic foot ulcers of 4 weeks in duration; ulcer of at least 1 cm2 in diameter and to the depth of subcutaneous tissue; healthy tissue at the ulcer; adequate arterial perfusion to heal; and ability to off-load the diabetic ulcer. The time to complete healing for PriMatrix was 38 days with 1.5 applications compared with 87 days with 2 applications for Apligraf. Although promising, additional study with a larger number of subjects is needed to compare the efficacy of PriMatrix with current SOC or advanced wound therapies.

Oasis Wound Matrix vs Regranex Gel

Niezgoda et al (2005) compared healing rates at 12 weeks for full-thickness diabetic foot ulcers treated with OASIS Wound Matrix (a porcine acellular wound care product) to Regranex Gel.42, This industry-sponsored, multicenter RCT was conducted at 9 outpatient wound care clinics and involved 73 patients with at least 1diabetic foot ulcer. Patients were randomized to receive either Oasis Wound Matrix (n=37) or Regranex Gel (n=36) and a secondary dressing. Wounds were cleansed and débrided, if needed, at a weekly visit. The maximum treatment period for each patient was 12 weeks. After 12 weeks, 18 (49%) Oasis-treated patients had complete wound closure compared with 10 (28%) Regranex-treated patients. Oasis treatment met the noninferiority margin but did not demonstrate that healing in the Oasis group was statistically superior (p=0.055). Post hoc subgroup analysis showed no significant difference in incidence of healing in patients with type 1 diabetes (33% vs 25%) but showed a significant improvement in patients with type 2 diabetes (63% vs 29%). There was also an increased healing of plantar ulcers in the Oasis group (52% vs 14%). These post hoc findings are considered hypothesis-generating. Additional study with a larger number of subjects is needed to compare the effect of Oasis treatment to current SOC.

Autologous Grafting on HYAFF Scaffolds

Uccioli et al(2011) reported a multicenter RCT of cultured expanded fibroblasts and keratinocytes grown on a HYAFF scaffold (benzyl ester of hyaluronic acid) compared with paraffin gauze for difficult diabetic foot ulcers.43, A total of 180 patients were randomized. At 12 weeks, complete ulcer healing was similar for the two groups (24% treated vs 21% controls). At 20 weeks, complete ulcer healing was achieved in

a similar proportion of the treatment group (50%) and the control group (43%, log-rank test = 0.344). Subgroup analysis, adjusted for baseline factors and possibly post-hoc, found a statistically significant benefit of treatment on dorsal ulcers but not plantar ulcers.

Section Summary: Bioengineered Skin Substitutes Other Than Apligraf, Dermagraft, AlloPatch, or Integra for Diabetic Lower-Extremity Ulcers

Results from a multicenter RCT showed some benefit of DermACELL that was primarily for the subgroup of patients who only required a single application of the ADM. Studies are needed to further define the population who might benefit from this treatment. Additional study with a larger number of subjects is needed to evaluate the effect of Graftjacket, TheraSkin, DermACELL, Cytal, PriMatrix, and Oasis Wound Matrix, compared with current SOC or other advanced wound therapies.

Lower-Extremity Ulcers due to Venous Insufficiency

Apligraf

Falanga et al (1998) reported on a multicenter randomized trial of Apligraf living cell therapy.44, A total of 293 patients with venous insufficiency and clinical signs of venous ulceration were randomized to compression therapy alone or to compression therapy and treatment with Apligraf. Apligraf was applied up to a maximum of 5 (mean, 3.3) times per patient during the initial 3 weeks. The primary end points were the percentage of patients with complete healing by 6 months after initiation of treatment and the time required for complete healing. At 6-month follow-up, the percentage of patients healed was higher with Apligraf (63% vs 49%), and the median time to complete wound closure was shorter (61 days vs 181 days). Treatment with Apligraf was superior to compression therapy in healing larger (>1000 mm2) and deeper ulcers and ulcers of more than 6 months in duration. There were no symptoms or signs of rejection, and the occurrence of adverse events was similar in both groups. This study was reviewed in a 2001 TEC Assessment, which concluded that Apligraf (Graftskin), in conjunction with good local wound care, met TEC criteria for the treatment of venous ulcers that fail to respond to conservative management.22,

Oasis Wound Matrix

Mostow et al (2005) reported on an industry-sponsored multicenter (12 sites) randomized trial that compared weekly treatment using Oasis Wound Matrix (xenogenic collagen scaffold from porcine small intestinal mucosa) with SOC in 120 patients who had chronic ulcers due to venous insufficiency that had not adequately responding to conventional therapy.45, Healing was assessed weekly for up to 12 weeks, with follow-up performed after 6 months to assess recurrence. After 12 weeks of treatment, there was a significant improvement in the percentage of wounds healed in the Oasis group (55% vs 34%). After adjusting for baseline ulcer size, patients in the Oasis group were 3 times more likely to heal than those in the group receiving SOC. Patients in the SOC group whose wounds did not heal by week 12 were allowed to cross over to Oasis treatment. None of the healed patients treated with Oasis wound matrix whowas seen for the 6-month follow-up experienced ulcer recurrence.

A research group in Europe has described 2 comparative studies of the Oasis matrix for mixed arteriovenous ulcers. In a 2007 quasirandomized study, Romanelli et al compared the efficacy of 2 extracellular matrix-based products, Oasis and Hyaloskin (extracellular matrix with hyaluronic acid).46, Fifty-four patients with mixed arteriovenous leg ulcers were assigned to the 2 arms based on order of entry into the study; 50 patients completed the study. Patients were followed twice weekly, and dressings changed more than once a week, only when necessary. After 16 weeks of treatment, complete wound closure was achieved in 82.6% of Oasis-treated ulcers compared with 46.2% of Hyaloskin-treated ulcers. Oasis treatment significantly increased the time to dressing change (mean, 6.4 days vs 2.4 days), reduced pain on a 10-point scale (3.7 vs 6.2), and improved patient comfort (2.5 vs 6.7).

In a 2010 trial, Romanelli et al compared Oasis with a moist wound dressing (SOC) in 23 patients with mixed arteriovenous ulcers and 27 patients with venous ulcers.47, The trial was described as randomized, but the method of randomization was not described. After the 8-week study period, patients were followed monthly for 6 months to assess wound closure. Complete wound closure was achieved in 80% of the Oasis-treated ulcers at 8 weeks compared with 65% of the SOC group. On average, Oasis-treated ulcers achieved complete healing in 5.4 weeks compared with 8.3 weeks for the SOC group. Treatment with Oasis also increased the time to dressing change (5.2 days vs 2.1 days) and the percentage of granulation tissue formed (65% vs 38%).

Subsection Summary: Apligraf or Oasis Wound Matrix for Lower-Extremity Ulcers due to Venous Insufficiency

RCTs have demonstrated the efficacy of Apligraf or Oasis Wound Matrix over SOC for lower-extremity ulcers due to venous insufficiency. Evidence is considered sufficient for these products.

Bioengineered Skin Substitutes Other Than Apligraf or Oasis Wound Matrix for Lower-Extremity Ulcers due to Venous Insufficiency

Dermagraft

Dermagraft living cell therapy has been approved by FDA for repair of diabetic foot ulcers. Use of Dermagraft for venous ulcers is an off-label indication. In 2013, Harding el al reported an open-label multicenter RCT that compared Dermagraft plus compression therapy (n=186) with compression therapy alone (n=180).48, The trial had numerous inclusion and exclusion criteria that restricted the population to patients who had nonhealing ulcers with compression therapy but had the capacity to heal. ITT analysis revealed no significant difference between the 2 groups in the primary outcome measure, the proportion of patients with completely healed ulcers by 12 weeks (34% Dermagraft vs 31% control). Prespecified subgroup analysis revealed a significant improvement in the percentage of wounds healed for ulcers of 12 months or less in duration (52% vs 37%) and for ulcers of 10 cm or less in diameter (47% vs 39%). There were no significant differences in the secondary outcomes of time to healing, complete healing by week 24, and percent reduction in ulcer area.

PriMatrix

In 2011, Karr published a retrospective comparison of PriMatrix (xenogenic ADM) and Apligraf in 28 venous stasis ulcers.41, The first 14 venous stasis ulcers matching the inclusion and exclusion criteria for each graft were compared. Criteria were venous stasis ulcers of 4 weeks in duration, at least 1 cm2 in diameter, and to a depth of subcutaneous tissue, with healthy tissue at the ulcer edge, adequate arterial perfusion to heal, and ability to tolerate compression therapy. The time to complete healing for PriMatrix was 32 days with 1.3 applications compared with 63 days with 1.7 applications for Apligraf. Although promising, additional study with a larger number of subjects is needed to assess the effect of PriMatrix treatment in compared with current SOC.

Section Summary: Bioengineered Skin Substitutes Other Than Apligraf or Oasis Wound Matrix for Lower-Extremity Ulcers due to Venous Insufficiency

In a moderately large RCT, Dermagraft was not shown to be more effective than controls in the primary or secondary end points for the entire population and was slightly more effective than controls (an 8%-15% increase in healing) only in subgroups of patients with ulcer duration of 12 months or less or wound diameter of 10 cm or less. Additional study with a larger number of subjects is needed to evaluate the effect of PriMatrix treatment compared with current SOC.

Dystrophic Epidermolysis Bullosa

OrCel was approved under an humanitarian device exemption (HDE) for use in patients with dystrophic epidermolysis bullosa undergoing hand reconstruction surgery, to close and heal wounds created by the surgery, including those at donor sites. HDE status has been withdrawn for Dermagraft for this indication.

Fivenson et al (2003) reported the off-label use of Apligraf in 5 patients with recessive dystrophic epidermolysis bullosa who underwent syndactyly release.49,

Section Summary: Dystrophic EpidermolysisBullosa

Dystrophic epidermolysis bullosa is a rare disorder. Because this is a rare disorder, it is unlikely that RCTs will be conducted to evaluate whether OrCel improves health outcomes for this condition. Therefore, the HDE for OrCel is considered sufficient.

Deep Dermal Burns

Epicel

One case series from 2000 has described the treatment of 30 severely burned patients with Epicel.50, The cultured epithelial autografts were applied to a mean of 37% of total body surface area (TBSA). Epicel achieved permanent coverage of a mean of 26% of TBSA, an area similar to that covered by conventional autografts (mean, 25%). Survival was 90% in these severely burned patients.

Integra Dermal Regeneration Template

A 2013 study compared Integra with split-thickness skin graft and with viscose cellulose sponge (Cellonex), using three 10´5 cm test sites on each of 10 burn patients.51, The surrounding burn area was covered with meshed autograft. Biopsies were taken from each site on days 3, 7, 14, and 21, and at months 3 and 12. The tissue samples were stained and examined for markers of inflammation and proliferation. The Vancouver Scar Scale was used to assess scars. At 12-month follow-up, the 3 methods resulted in similar clinical appearance, along with similar histologic and immunohistochemical findings.

Branski et al (2007) reported on a randomized trial that compared Integra with a standard autograft-allograft technique in 20 children with an average burn size of 73% TBSA (71% full-thickness burns).52, Once vascularized (about 14-21 days), the Silastic epidermis was stripped and replaced with thin (0.05-0.13 mm) epidermal autograft. There were no significant differences between the Integra group and controls in burn size (70% vs 74% TBSA), mortality (40% vs 30%), and hospital length of stay (41 vs 39 days), all respectively. Long-term follow-up revealed a significant increase in bone mineral content and density (24 months) and improved scarring in terms of height, thickness, vascularity, and pigmentation (at 12 months and 18-24 months) in the Integra group. No differences were observed between groups in the time to first reconstructive procedure, cumulative reconstructive procedures required during 2 years, and cumulative operating room time required for these procedures. The authors concluded that Integra can be used for immediate wound coverage in children with severe burns without the associated risks of cadaver skin.

Heimbach et al (2003) reported on a multicenter (13 U.S. burn care facilities) postapproval study involving 222 burn injury patients (36.5% TBSA; range, 1%-95%) who were treated with Integra Dermal Regeneration Template.53, Within 2 to 3 weeks, the dermal layer regenerated, and a thin epidermal autograft was placed over the wound. The incidence of infection was 16.3%. Mean take rate (absence of graft failure) of Integra was 76.2%; the median take rate was 98%. The mean take rate of epidermal autograft placed over Integra was 87.7%; the median take rate was 95%.

TransCyte

TransCyte is no longer commercially available.

Earlier studies included a 2001 report by Lukish et al that found improved healing in 20 consecutive cases of pediatric burns greater than 7% TBSA that underwent wound closure using TransCyte compared to the previous 20 consecutive burn cases greater than 7% TBSA that received standard therapy.54, In 2006, Amani et alfound significant improvement in healing in 110 consecutive patients who had deep partial-thickness burns treated with TransCyte as compared to results from the American Burn Association Patient Registry for similar burns.55,

Section Summary: Deep Dermal Burns

Epicel isFDA-approved under an HDE for the treatment of deep dermal or full-thickness burns comprising a TBSA of 30% or more, with patient survival of 90%. Integra Dermal Regeneration Template has been compared with autograft in a within-subject study and with autograft-allograft in a small RCT with 10 patients per group. Outcomes are at least as good as with autograft or allograft, with a reduction in scarring and without risks associated with cadaver skin. This product has also been studied in a large series with over 222 burn patients, showing a take rate of 76% and with a take rate of epidermal autograft placed over Integra of 87.7%.

Other Indications

Punch Biopsy Wounds

Baldursson et al (2015) reported a double-blinded RCT with 81 patients (162 punch biopsy wounds) that compared Kerecis Omega3 Wound (derived from fish skin) with Oasis SIS ECM (porcine small intestinal submucosa extracellular matrix).56, The primary outcome (the percentage of wounds healed at 28 days) was similar for the fish skin ADM (95%) and the porcine SIS ECM (96.3%). The rate of healing was faster with Kerecis Omega3 (p=0.041). At 21 days, 72.5% of the fish skin ADM group had healed compared with 56% of the porcine SIS ECM group. Interpretation of this study is limited because it did not include an accepted control condition for this indication.

Split-Thickness Donor Sites

There is limited evidence to support the efficacy of OrCel compared with SOC for the treatment of split-thickness donor sites in burn patients. In 2003, Stillet alexamined the safety and efficacy of bilayered OrCel to facilitate wound closure of split-thickness donor sites in 82 severely burned patients.57, Each patient had 2 designated donor sites that were randomized to a single treatment of OrCel or standard dressing (Biobrane-L). The healing time for OrCel sites was significantly shorter than for sites treated with a standard dressing, enabling earlier recropping. OrCel sites also exhibited a nonsignificant trend for reduced scarring. Additional studies are needed to evaluate the effect of this product on health outcomes.

Miscellaneous

In addition to indications previously reviewed, off-label uses of bioengineered skin substitutes have included pressure ulcers, inflammatory ulcers (eg, pyoderma gangrenosum, vasculitis), scleroderma digital ulcers, postkeloid removal wounds, genetic conditions, and variety of other conditions.58 Products that have been FDA-approved or -cleared for one indication (eg, lower-extremity ulcers) have also been used off-label in place of other FDA-approved or -cleared products (eg, for burns).59, No controlled trials were identified for these indications.

Summary of Evidence

Breast Reconstruction

For individuals who are undergoing breast reconstruction who receive allogeneic ADM products, the evidence incudes RCTs and systematic reviews. Relevant outcomes are symptoms, morbid events, functional outcomes, quality of life, and treatment-related morbidity. A systematic review found no difference in overall complication rates with ADM allograft compared with standard procedures for breast reconstruction. Reconstructions with ADM have been reported to have higher seroma, infection, and necrosis rates than reconstructions without ADM. However, capsular contracture and malposition of implants may be reduced. Thus, in cases where there is limited tissue coverage, the available evidence may inform patient decision making about reconstruction options. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

Tendon Repair

For individuals who are undergoing tendon repair who receive Graftjacket, the evidence incudes an RCT. Relevant outcomes are symptoms, morbid events, functional outcomes, quality of life, and treatment-related morbidity. The RCT identified found improved outcomes with the Graftjacket ADM allograft for rotator cuff repair. Although these results were positive, additional study with a larger number of patients is needed to evaluate the consistency of the effect. The evidence is insufficient to determine the effects of the technology on health outcomes.

Surgical Repair of Hernias or Parastomal Reinforcement

For individuals who are undergoing surgical repair of hernias or parastomal reinforcement who receive acellular collagen-based scaffolds, the evidence incudesRCTs. Relevant outcomes are symptoms, morbid events, functional outcomes, quality of life, and treatment-related morbidity. Several comparative studies including RCTs have shown no difference in outcomes between tissue-engineered skin substitutes and either standard synthetic mesh or no reinforcement. The evidence is sufficient to determine that the technology is unlikely to improve the net health outcome.

Diabetic Lower-Extremity Ulcers

For individuals who have diabetic lower-extremity ulcers who receive AlloPatch, Apligraf, Dermagraft, or Integra, the evidence includes RCTs. Relevant outcomes are disease-specific survival, symptoms, change in disease status, morbid events, and quality of life. RCTs have demonstrated the efficacy of AlloPatch (reticular ADM), Apligraf and Dermagraft (living cell therapy), and Integra (biosynthetic) over the standard of care. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

For individuals who have diabetic lower-extremity ulcers who receive ADM products other than AlloPatch, Apligraf, Dermagraft, or Integra, the evidence includes RCTs. Relevant outcomes are disease-specific survival, symptoms, change in disease status, morbid events, and quality of life. Results from a multicenter RCT showed some benefit of DermACELL that was primarily for the subgroup of patients who only required a single application of the ADM. Studies are needed to further define the population who might benefit from this treatment. Additional study with a larger number of subjects is needed to evaluate the effect of Graftjacket, TheraSkin, DermACELL, Cytal, PriMatrix, and Oasis Wound Matrix, compared with current SOC or other advanced wound therapies. The evidence is insufficient to determine the effects of the technology on health outcomes.

Lower-Extremity Ulcers due to Venous Insufficiency

For individuals who have lower-extremity ulcers due to venous insufficiency who receive Apligraf or Oasis Wound Matrix, the evidence includes RCTs. Relevant outcomes are disease-specific survival, symptoms, change in disease status, morbid events, and quality of life. RCTs have demonstrated the efficacy of Apligraf living cell therapy and xenogenic Oasis Wound Matrix over the standard of care. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

For individuals who have lower-extremity ulcers due to venous insufficiency who receive bioengineered skin substitutes other than Apligraf or Oasis Wound Matrix, the evidence includes RCTs. Relevant outcomes are disease-specific survival, symptoms, change in disease status, morbid events, and quality of life. In a moderately large RCT, Dermagraft was not shown to be more effective than controls for the primary or secondary end points in the entire population and was only slightly more effective than controls (an 8%-15% increase in healing) in subgroups of patients with ulcer durations of 12 months or less or size of 10 cm or less. Additional study with a larger number of subjects is needed to evaluate the effect of the xenogenic PriMatrix skin substitute vs the current standard of care. The evidence is insufficient to determine the effects of the technology on health outcomes.

Dystrophic Epidermolysis Bullosa

For individuals who have dystrophic epidermolysis bullosa who receive OrCel, the evidence includes case series. Relevant outcomes are symptoms, change in disease status, morbid events, and quality of life. OrCel was approved under a humanitarian drug exemption for use in patients with dystrophic epidermolysis bullosa undergoing hand reconstruction surgery, to close and heal wounds created by the surgery, including those at donor sites. Outcomes have been reported in small series (eg, 5 patients). The evidence is insufficient to determine the effects of the technology on health outcomes.

Deep Dermal Burns

For individuals who have deep dermal burns who receive bioengineered skin substitutes (ie, Epicel, Integra Dermal Regeneration Template), the evidence includes RCTs. Relevant outcomes are disease-specific survival, symptoms, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related morbidity. Overall, few skin substitutes have been approved, and the evidence is limited for each product. Epicel (living cell therapy) has received FDA approval under a humanitarian device exemption for the treatment of deep dermal or full-thickness burns comprising a total body surface area of 30% or more. Comparative studies have demonstrated improved outcomes for biosynthetic skin substitute Integra Dermal Regeneration Template for the treatment of burns. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

SUPPLEMENTAL INFORMATION

Clinical Input From Physician Specialty Societies and Academic Medical Centers

While the various physician specialty societies and academic medical centers may collaborate with and make recommendations during this process, through the provision of appropriate reviewers, input received does not represent an endorsement or position statement by the physician specialty societies or academic medical centers, unless otherwise noted.

2016 Input

In response to requests, input was received from 2 physician specialty societies and 3 academic medical centers while this policy was under review in 2016. Input was requested on the equivalency of products within the categories of amniotic membrane, living cell therapies, and biosynthetic skin substitutes for the treatment of diabetic foot ulcers and nonocular burns (biosynthetic only). Input on the equivalency of products within these categories was mixed.

2014 Input

In response to requests, input was received from 3 physician specialty societies and 4 academic medical centers while this policy was under review in 2014. In addition to questions on medical necessity for different indications, input was specifically requested on the equivalency of products within the different categories (eg, acellular dermal matrix [ADM], living cell therapy, xenogeneic collagen scaffold, amniotic membrane). Five reviewers addressed the use of ADM products for breast reconstruction and most considered the various ADM products (AlloDerm, AlloMax, DermaMatrix, FlexHD, Graftjacket) to have similar outcomes when used for breast reconstructive surgery, although differences in firmness and stretch of the products were noted. Six reviewers addressed questions on bioengineered skinand soft tissue substitutes for diabetic and venous lower-extremity ulcers. Responses were mixed, although most reviewers considered living cell therapies to be equivalent for these indications. Most reviewers did not consider xenogeneic ADM products (eg, PriMatrix) or amniotic membrane (eg, EpiFix) to be medically necessary for any indication.

2012 Input

In response to requests, input was received from 3 physician specialty societies and 2 academic medical centers while this policy was under review in 2012. Most reviewers supported the indications and products described in this policy. Input was requested on the use of an interpositional spacer after parotidectomy. Support for this indication was mixed. Some reviewers suggested use of other products and/or additional indications; however, the input on these products/indications was not uniform. Reviewers provided references for the additional indications; these were subsequently reviewed.

2009 Input

In response to requests, input was received from 1 physician specialty society (2 physicians) and 1 academic medical center while this policy was under review in 2009. All reviewers indicated that on use of AlloDerm in breast reconstruction surgery should be available for use during breast reconstructive surgery.

Practice Guidelines and Position Statements

American Society of Plastic Surgeons and Wound Healing Society

A literature review for the 2013 guidelines from the American Society of Plastic Surgeons (ASPS) found that use of ADM, although increasingly common in postmastectomy expander/implant breast reconstruction, can result in increased risk of complications in the presence of certain risk factors.60, ASPS noted that cellular dermal matrix is currently used to increase soft tissue coverage, support the implant pocket, improve contour, and reduce pain with expansion. However, evidence to support these improved surgical outcomes are limited. Some evidence has suggested that use of ADM is associated with increased postoperative complications, specifically related to infection and seroma. Overall, ASPS found that evidence on ADM products in postmastectomy expander/implant breast reconstruction was varied and conflicting, and gave a grade C recommendation based on level III evidence that surgeons should evaluate each clinical case individually and objectively determine the use of ADM.

National Institute for Health and Care Excellence

In 2016, the National Institute for Health and Care Excellence updated its guidance on the prevention and management of diabetic foot problems.61, The Institute recommended that clinicians “consider dermal or skin substitutes as an adjunct to standard care when treating diabetic foot ulcers, only when healing has not progressed and on the advice of the multidisciplinary foot care service.”

Infectious Diseases Society of America

The 2012 guidelines from the Infectious Diseases Society of America stated that, for selected diabetic foot wounds that are slow to heal, clinicians might consider using bioengineered skin equivalents (weak recommendation, moderate evidence), growth factors (weak, moderate), granulocyte colony-stimulating factors (weak, moderate), hyperbaric oxygen therapy (strong, moderate), or negative pressure wound therapy (weak, low).62, It was emphasized that none of these measures had been shown to improve the resolution of infection and that they were expensive, not universally available, mighty require consultation with experts, and reports supporting their utility were mostly flawed.

U.S. Preventive Services Task Force Recommendations

Not applicable.

Ongoing and Unpublished Clinical Trials

Some currently unpublished trials that might influence this review are listed in Table 6.

Table 6. Summary of Key Trials
NCT No.Trial NamePlanned EnrollmentCompletion Date
Ongoing 
 
 
NCT01987700aMulti-Center Study To Examine The Use Of Flex HD® And Strattice In The Repair Of Large Abdominal Wall Hernias
120
Oct 2017
(ongoing)
NCT02587403aA Randomized, Prospective Study Comparing Fortiva™ Porcine Dermis vs. Strattice™Reconstructive Tissue Matrix in Patients Undergoing Complex Open Primary Ventral Hernia Repair
120
Oct 2019
NCT02322554The Registry of Cellular and Tissue Based Therapies for Chronic Wounds and Ulcers
50,000
Jan 2020
Unpublished 
 
 
NCT01970163aA Multicenter, Randomized, Controlled, Open Label Trial of DermACELL in Subjects With Chronic Wounds of the Lower Extremities
202
Apr 2016
(completed)
NCT: national clinical trial.

a Denotes industry-sponsored or cosponsored trial.]
________________________________________________________________________________________

Horizon BCBSNJ Medical Policy Development Process:

This Horizon BCBSNJ Medical Policy (the “Medical Policy”) has been developed by Horizon BCBSNJ’s Medical Policy Committee (the “Committee”) consistent with generally accepted standards of medical practice, and reflects Horizon BCBSNJ’s view of the subject health care services, supplies or procedures, and in what circumstances they are deemed to be medically necessary or experimental/ investigational in nature. This Medical Policy also considers whether and to what degree the subject health care services, supplies or procedures are clinically appropriate, in terms of type, frequency, extent, site and duration and if they are considered effective for the illnesses, injuries or diseases discussed. Where relevant, this Medical Policy considers whether the subject health care services, supplies or procedures are being requested primarily for the convenience of the covered person or the health care provider. It may also consider whether the services, supplies or procedures are more costly than an alternative service or sequence of services, supplies or procedures that are at least as likely to produce equivalent therapeutic or diagnostic results as to the diagnosis or treatment of the relevant illness, injury or disease. In reaching its conclusion regarding what it considers to be the generally accepted standards of medical practice, the Committee reviews and considers the following: all credible scientific evidence published in peer-reviewed medical literature generally recognized by the relevant medical community, physician and health care provider specialty society recommendations, the views of physicians and health care providers practicing in relevant clinical areas (including, but not limited to, the prevailing opinion within the appropriate specialty) and any other relevant factor as determined by applicable State and Federal laws and regulations.

___________________________________________________________________________________________________________________________

Index:
Bioengineered Skin and Soft Tissue Substitutes
Bio-Engineered Skin and Soft Tissue Substitutes
ACell® UBM Hydated Wound Dressing
ACell® UBM Lyophilized Wound Dressing
AlloDerm®
AlloMax™
AlloMend®
AlloPatch®
AlloSkin™
AlloSkin™ RT
Alphaplex™ with MariGen Omega3™
Aongen™ Collagen Matrix
Apligraf®
Architect® ECM, PX, FX
ArthroFlex™ (FlexGraft)
Atlas Wound Matrix
Avagen Wound Dressing
Avaulta Plus™
AxoGuard® Nerve Protector (AxoGen)
CellerateRX®
CollaCare®
CollaCare® Dental
Collagen Sponge (Innocoll)
Collagen Wound Dressing (Oasis Research)
CollaGUARD®
CollaMend™
CollaSorb™
CollaWound™
Collexa®
Collieva®
Conexa™
Coreleader Colla-Pad
CorMatrix®
CRXa™
Cymetra®
Cytal™ (previously MatriStem®)
Dermadapt™ Wound Dressing
Dermagraft®
DermaMatrix™
DermaPure™
DermaSpan™
DressSkin
Durepair Regeneration Matrix®
Endoform Dermal Template™
ENDURAgen™
Epicel®
Excellagen
ExpressGraft™
E-Z Derm™
FlexHD®
FlexiGraft®
FortaDerm™ Wound Dressing
GammaGraft
Graftjacket®
Graftjacket® Xpress
HA Absorbent Wound Dressing
Helicoll
Hyalomatrix®
Hyalomatrix® PA
hMatrix®
Integra Dermal Regeneration Template
Integra™ Flowable Wound Matrix
Integra™ Bilayer Wound Matrix
Integra™ Omnigraft
Jaloskin®
Kerecis™
MariGen™
MatriDerm®
MatriStem® Burn Matrix
MatriStem® Micromatrix
MatriStem® Wound Matrix
Matrix Collagen Wound Dressing
Matrix HD™
MediHoney®
Mediskin®
MemoDerm™
Microderm® biologic wound matrix
NeoForm™
NuCel
Oasis® Burn Matrix
Oasis® Ultra
Oasis™ Wound Matrix
Omega3™
OrCel™
Pelvicol®/PelviSoft®
Permacol™
PriMatrix™
Primatrix™ Dermal Repair Scaffold
PuraPly™ Wound Matrix (previously FortaDerm™)
PuraPly™ AM (Antimicrobial Wound Matrix)
Puros® Dermis
RegenePro™
Repliform®
Repriza™
SIS Wound Dressing II
SS Matrix™
Stimulen™ Collagen
StrataGraft
Strattice™
Suprathel®
SurgiMend®
Talymed®
TenoGlide™
TenSIX™ Acellular Dermal Matrix
TheraForm™ Standard/Sheet
TheraSkin®Unite™
TissueMend
TruSkin™
Unite® Biomatrix
Veritas® Collagen Matrix
XCM Biologic® Tissue Matrix
XenMatrix™ AB

References:
1. Davila AA, Seth AK, Wang E, et al. Human acellular dermis versus submuscular tissue expander breast reconstruction: a multivariate analysis of short-term complications. Archives of plastic surgery. Jan 2013;40(1):19-27. PMID 23362476

2. Lee KT, Mun GH. Updated evidence of acellular dermal matrix use for implant-based breast reconstruction: a meta-analysis. Ann Surg Oncol. Feb 2016;23(2):600-610. PMID 26438439

3. McCarthy CM, Lee CN, Halvorson EG, et al. The use of acellular dermal matrices in two-stage expander/implant reconstruction: a multicenter, blinded, randomized controlled trial. Plast Reconstr Surg. Nov 2012;130(5 Suppl 2):57S-66S. PMID 23096987

4. Hinchcliff KM, Orbay H, Busse BK, Charvet H, Kaur M, Sahar DE. Comparison of two cadaveric acellular dermal matrices for immediate breast reconstruction: A prospective randomized trial. Journal of plastic, reconstructive & aesthetic surgery : JPRAS. May 2017;70(5):568-576. PMID 28341592

5. Mendenhall SD, Anderson LA, Ying J, Boucher KM, Neumayer LA, Agarwal JP. The BREASTrial Stage II: ADM breast reconstruction outcomes from definitive reconstruction to 3 months postoperative. Plastic and reconstructive surgery Global open. Jan 2017;5(1):e1209. PMID 28203509

6. Liu DZ, Mathes DW, Neligan PC, Said HK, Louie O. Comparison of outcomes using AlloDerm versus FlexHD for implant-based breast reconstruction. Ann Plast Surg. May 2014;72(5):503-507. PMID 23636114

7. Chang EI, Liu J. Prospective unbiased experience with three acellular dermal matrices in breast reconstruction. J Surg Oncol. Sep 2017;116(3):365-370. PMID 28444764

8. Pittman TA, Fan KL, Knapp A, Frantz S, Spear SL. Comparison of Different Acellular Dermal Matrices in Breast Reconstruction: The 50/50 Study. Plast Reconstr Surg. Mar 2017;139(3):521-528. PMID 28234811

9. Dikmans RE, Negenborn VL, Bouman MB, et al. Two-stage implant-based breast reconstruction compared with immediate one-stage implant-based breast reconstruction augmented with an acellular dermal matrix: an open-label, phase 4, multicentre, randomised, controlled trial. The Lancet Oncology. Feb 2017;18(2):251-258. PMID 28012977

10. Barber FA, Burns JP, Deutsch A, Labbe MR, Litchfield RB. A prospective, randomized evaluation of acellular human dermal matrix augmentation for arthroscopic rotator cuff repair. Arthroscopy. Jan 2012;28(1):8-15. PMID 21978432

11. Bellows CF, Smith A, Malsbury J, Helton WS. Repair of incisional hernias with biological prosthesis: a systematic review of current evidence. Am J Surg. Jan 2013;205(1):85-101. PMID 22867726

12. Espinosa-de-los-Monteros A, de la Torre JI, Marrero I, Andrades P, Davis MR, Vasconez LO. Utilization of human cadaveric acellular dermis for abdominal hernia reconstruction. Ann Plast Surg. Mar 2007;58(3):264-267. PMID 17471129

13. Gupta A, Zahriya K, Mullens PL, Salmassi S, Keshishian A. Ventral herniorrhaphy: experience with two different biosynthetic mesh materials, Surgisis and Alloderm. Hernia : the journal of hernias and abdominal wall surgery. Oct 2006;10(5):419-425. PMID 16924395

14. Bochicchio GV, De Castro GP, Bochicchio KM, Weeks J, Rodriguez E, Scalea TM. Comparison study of acellular dermal matrices in complicated hernia surgery. J Am Coll Surg. Oct 2013;217(4):606-613. PMID 23973102

15. Roth JS, Zachem A, Plymale MA, Davenport DL. Complex ventral hernia repair with acellular dermal matrices: clinical and quality of life outcomes. Am Surg. Feb 1 2017;83(2):141-147. PMID 28228200

16. Bellows CF, Shadduck P, Helton WS, Martindale R, Stouch BC, Fitzgibbons R. Early report of a randomized comparative clinical trial of Strattice reconstructive tissue matrix to lightweight synthetic mesh in the repair of inguinal hernias. Hernia : the journal of hernias and abdominal wall surgery. Apr 2014;18(2):221-230. PMID 23543334

17. Fleshman JW, Beck DE, Hyman N, et al. A prospective, multicenter, randomized, controlled study of non-cross-linked porcine acellular dermal matrix fascial sublay for parastomal reinforcement in patients undergoing surgery for permanent abdominal wall ostomies. Dis Colon Rectum. May 2014;57(5):623-631. PMID 24819103

18. Santema TB, Poyck PP, Ubbink DT. Skin grafting and tissue replacement for treating foot ulcers in people with diabetes. Cochrane database of systematic reviews. Feb 11 2016;2:CD011255. PMID 26866804

19. Martinson M, Martinson N. A comparative analysis of skin substitutes used in the management of diabetic foot ulcers. J Wound Care. Oct 2016;25(Sup10):S8-S17. PMID 27681811

20. Guo X, Mu D, Gao F. Efficacy and safety of acellular dermal matrix in diabetic foot ulcer treatment: A systematic review and meta-analysis. International journal of surgery (London, England). Apr 2017;40:1-7. PMID 28232031

21. Veves A, Falanga V, Armstrong DG, Sabolinski ML. Graftskin, a human skin equivalent, is effective in the management of noninfected neuropathic diabetic foot ulcers: a prospective randomized multicenter clinical trial. Diabetes Care. Feb 2001;24(2):290-295. PMID 11213881

22. Blue Cross and Blue Shield Technology Evaluation Center (TEC). Graftskin for the treatment of skin ulcers. TEC Assessments. 2001;Volume 16:Tab 12. PMID

23. Steinberg JS, Edmonds M, Hurley DP, Jr., King WN. Confirmatory data from EU study supports Apligraf for the treatment of neuropathic diabetic foot ulcers. J Am Podiatr Med Assoc. Jan-Feb 2010;100(1):73-77. PMID 20093548

24. Kirsner RS, Warriner R, Michela M, Stasik L, Freeman K. Advanced biological therapies for diabetic foot ulcers. Arch Dermatol. Aug 2010;146(8):857-862. PMID 20713816

25. Marston WA, Hanft J, Norwood P, Pollak R, Dermagraft Diabetic Foot Ulcer Study G. The efficacy and safety of Dermagraft in improving the healing of chronic diabetic foot ulcers: results of a prospective randomized trial. Diabetes Care. Jun 2003;26(6):1701-1705. PMID 12766097

26. Frykberg RG, Marston WA, Cardinal M. The incidence of lower-extremity amputation and bone resection in diabetic foot ulcer patients treated with a human fibroblast-derived dermal substitute. Advances in skin & wound care. Jan 2015;28(1):17-20. PMID 25407083

27. Zelen CM, Orgill DP, Serena T, et al. A prospective, randomised, controlled, multicentre clinical trial examining healing rates, safety and cost to closure of an acellular reticular allogenic human dermis versus standard of care in the treatment of chronic diabetic foot ulcers. International wound journal. Apr 2017;14(2):307-315. PMID 27073000

28. Zelen CM, Orgill DP, Serena TE, et al. An aseptically processed, acellular, reticular, allogenic human dermis improves healing in diabetic foot ulcers: A prospective, randomised, controlled, multicentre follow-up trial. International wound journal. Oct 2018;15(5):731-739. PMID 29682897

29. Driver VR, Lavery LA, Reyzelman AM, et al. A clinical trial of Integra Template for diabetic foot ulcer treatment. Wound Repair Regen. Nov 12 2015;23(6):891-900. PMID 26297933

30. Campitiello F, Mancone M, Della Corte A, Guerniero R, Canonico S. To evaluate the efficacy of an acellular Flowable matrix in comparison with a wet dressing for the treatment of patients with diabetic foot ulcers: a randomized clinical trial. Updates in surgery. Dec 2017;69(4):523-529. PMID 28497218

31. Brigido SA, Boc SF, Lopez RC. Effective management of major lower extremity wounds using an acellular regenerative tissue matrix: a pilot study. Orthopedics. Jan 2004;27(1 Suppl):s145-149. PMID 14763548

32. Reyzelman A, Crews RT, Moore JC, et al. Clinical effectiveness of an acellular dermal regenerative tissue matrix compared to standard wound management in healing diabetic foot ulcers: a prospective, randomised, multicentre study. International wound journal. Jun 2009;6(3):196-208. PMID 19368581

33. Reyzelman AM, Bazarov I. Human acellular dermal wound matrix for treatment of DFU: literature review and analysis. J Wound Care. Mar 2015;24(3):128; 129-134. PMID 25764957

34. Brigido SA. The use of an acellular dermal regenerative tissue matrix in the treatment of lower extremity wounds: a prospective 16-week pilot study. International wound journal. Sep 2006;3(3):181-187. PMID 16984575

35. Walters J, Cazzell S, Pham H, Vayser D, Reyzelman A. Healing rates in a multicenter assessment of a sterile, room temperature, acellular dermal matrix versus conventional care wound management and an active comparator in the treatment of full-thickness diabetic foot ulcers. Eplasty. Mar 2016;16:e10. PMID 26933467

36. Cazzell S, Vayser D, Pham H, et al. A randomized clinical trial of a human acellular dermal matrix demonstrated superior healing rates for chronic diabetic foot ulcers over conventional care and an active acellular dermal matrix comparator. Wound Repair Regen. May 2017;25(3):483-497. PMID 28544150

37. Sanders L, Landsman AS, Landsman A, et al. A prospective, multicenter, randomized, controlled clinical trial comparing a bioengineered skin substitute to a human skin allograft. Ostomy Wound Manage. Sep 2014;60(9):26-38. PMID 25211605

38. DiDomenico L, Landsman AR, Emch KJ, Landsman A. A prospective comparison of diabetic foot ulcers treated with either a cryopreserved skin allograft or a bioengineered skin substitute. Wounds : a compendium of clinical research and practice. Jul 2011;23(7):184-189. PMID 25879172

39. Frykberg RG, Cazzell SM, Arroyo-Rivera J, et al. Evaluation of tissue engineering products for the management of neuropathic diabetic foot ulcers: an interim analysis. J Wound Care. Jul 2016;25 Suppl 7:S18-25. PMID 27410467

40. Kavros SJ, Dutra T, Gonzalez-Cruz R, et al. The use of PriMatrix, a fetal bovine acellular dermal matrix, in healing chronic diabetic foot ulcers: a prospective multicenter study. Advances in skin & wound care. Aug 2014;27(8):356-362. PMID 25033310

41. Karr JC. Retrospective comparison of diabetic foot ulcer and venous stasis ulcer healing outcome between a dermal repair scaffold (PriMatrix) and a bilayered living cell therapy (Apligraf). Advances in skin & wound care. Mar 2011;24(3):119-125. PMID 21326023

42. Niezgoda JA, Van Gils CC, Frykberg RG, Hodde JP. Randomized clinical trial comparing OASIS Wound Matrix to Regranex Gel for diabetic ulcers. Advances in skin & wound care. Jun 2005;18(5 Pt 1):258-266. PMID 15942317

43. Uccioli L, Giurato L, Ruotolo V, et al. Two-step autologous grafting using HYAFF scaffolds in treating difficult diabetic foot ulcers: results of a multicenter, randomized controlled clinical trial with long-term follow-up. The international journal of lower extremity wounds. Jun 2011;10(2):80-85. PMID 21693443

44. Falanga V, Margolis D, Alvarez O, et al. Rapid healing of venous ulcers and lack of clinical rejection with an allogeneic cultured human skin equivalent. Human Skin Equivalent Investigators Group. Arch Dermatol. Mar 1998;134(3):293-300. PMID 9521027

45. Mostow EN, Haraway GD, Dalsing M, Hodde JP, King D. Effectiveness of an extracellular matrix graft (OASIS Wound Matrix) in the treatment of chronic leg ulcers: a randomized clinical trial. J Vasc Surg. May 2005;41(5):837-843. PMID 15886669

46. Romanelli M, Dini V, Bertone M, Barbanera S, Brilli C. OASIS wound matrix versus Hyaloskin in the treatment of difficult-to-heal wounds of mixed arterial/venous aetiology. International wound journal. Mar 2007;4(1):3-7. PMID 17425543

47. Romanelli M, Dini V, Bertone MS. Randomized comparison of OASIS wound matrix versus moist wound dressing in the treatment of difficult-to-heal wounds of mixed arterial/venous etiology. Advances in skin & wound care. Jan 2010;23(1):34-38. PMID 20101114

48. Harding K, Sumner M, Cardinal M. A prospective, multicentre, randomised controlled study of human fibroblast-derived dermal substitute (Dermagraft) in patients with venous leg ulcers. International wound journal. Apr 2013;10(2):132-137. PMID 23506344

49. Fivenson DP, Scherschun L, Cohen LV. Apligraf in the treatment of severe mitten deformity associated with recessive dystrophic epidermolysis bullosa. Plast Reconstr Surg. Aug 2003;112(2):584-588. PMID 12900618

50. Carsin H, Ainaud P, Le Bever H, et al. Cultured epithelial autografts in extensive burn coverage of severely traumatized patients: a five year single-center experience with 30 patients. Burns. Jun 2000;26(4):379-387. PMID 10751706

51. Lagus H, Sarlomo-Rikala M, Bohling T, Vuola J. Prospective study on burns treated with Integra(R), a cellulose sponge and split thickness skin graft: comparative clinical and histological study--randomized controlled trial. Burns. Dec 2013;39(8):1577-1587. PMID 23880091

52. Branski LK, Herndon DN, Pereira C, et al. Longitudinal assessment of Integra in primary burn management: a randomized pediatric clinical trial. Crit Care Med. Nov 2007;35(11):2615-2623. PMID 17828040

53. Heimbach DM, Warden GD, Luterman A, et al. Multicenter postapproval clinical trial of Integra dermal regeneration template for burn treatment. J Burn Care Rehabil. Jan-Feb 2003;24(1):42-48. PMID 12543990

54. Lukish JR, Eichelberger MR, Newman KD, et al. The use of a bioactive skin substitute decreases length of stay for pediatric burn patients. J Pediatr Surg. Aug 2001;36(8):1118-1121. PMID 11479839

55. Amani H, Dougherty WR, Blome-Eberwein S. Use of Transcyte and dermabrasion to treat burns reduces length of stay in burns of all size and etiology. Burns. Nov 2006;32(7):828-832. PMID 16997480

56. Baldursson BT, Kjartansson H, Konradsdottir F, Gudnason P, Sigurjonsson GF, Lund SH. Healing rate and autoimmune safety of full-thickness wounds treated with fish skin acellular dermal matrix versus porcine small-intestine submucosa: a noninferiority study. The international journal of lower extremity wounds. Mar 2015;14(1):37-43. PMID 25759413

57. Still J, Glat P, Silverstein P, Griswold J, Mozingo D. The use of a collagen sponge/living cell composite material to treat donor sites in burn patients. Burns. Dec 2003;29(8):837-841. PMID 14636761

58. Lazic T, Falanga V. Bioengineered skin constructs and their use in wound healing. Plast Reconstr Surg. Jan 2011;127 Suppl 1:75S-90S. PMID 21200276

59. Saffle JR. Closure of the excised burn wound: temporary skin substitutes. Clin Plast Surg. Oct 2009;36(4):627-641. PMID 19793557

60. American Society of Plastic Surgeons. Evidence-Based Clinical Practice Guideline: Breast Reconstruction with Expanders and Implants. 2013; http://www.plasticsurgery.org/Documents/medical-professionals/health-policy/evidence-practice/breast-reconstruction-expanders-with-implants-guidelines.pdf. Accessed January 2, 2018.

61. National Institute for Health and Care Excellence (NICE). Diabetic Foot Problems: Prevention and Management [NG19]. 2016; https://www.nice.org.uk/guidance/ng19/evidence. Accessed January 2, 2018.

62. Lipsky BA, Berendt AR, Cornia PB, et al. 2012 Infectious Diseases Society of America clinical practice guideline for the diagnosis and treatment of diabetic foot infections. Clin Infect Dis. Jun 2012;54(12):e132-173. PMID 22619242

63. Centers for Medicare & Medicaid Services (CMS). National Coverage Determination (NCD) for Porcine Skin and Gradient Pressure Dressings (270.5). n.d.; https://www.cms.gov/medicare-coverage-database/details/ncd-details.aspx?NCDId=139&ncdver=1&bc=AgAAQAAAAAAA&. Accessed January 2, 2018.

64. Centers for Medicare & Medicaid Services (CMS). Fact Sheet: CMS issues hospital outpatient department and ambulatory surgical center policy and payment changes for 2014. 2013; http://www.cms.gov/Newsroom/MediaReleaseDatabase/Fact-Sheets/2013-Fact-Sheets-Items/2013-11-27-3.html. Accessed January 2, 2018.

Codes:
(The list of codes is not intended to be all-inclusive and is included below for informational purposes only. Inclusion or exclusion of a procedure, diagnosis, drug or device code(s) does not constitute or imply authorization, certification, approval, offer of coverage or guarantee of payment.)

CPT*

    15040-15278
    15777
HCPCS
    Q4100
    Q4101
    Q4102
    Q4103
    Q4104
    Q4105
    Q4106
    Q4107
    Q4108
    Q4110
    Q4111
    Q4112
    Q4113
    Q4114
    Q4115
    Q4116
    Q4117
    Q4118
    Q4121
    Q4122
    Q4123
    Q4124
    Q4125
    Q4126
    Q4127
    Q4128
    Q4130
    Q4134
    Q4135
    Q4136
    Q4141
    Q4142
    Q4143
    Q4146
    Q4147
    Q4149
    Q4152
    Q4158
    Q4161
    Q4164
    Q4165
    Q4166
    Q4167
    Q4172
    Q4175
    Q4176
    Q4177
    Q4178
    Q4179
    Q4180
    Q4181
    Q4182

* CPT only copyright 2020 American Medical Association. All rights reserved. CPT is a registered trademark of the American Medical Association.

_________________________________________________________________________________________

Medical policies can be highly technical and are designed for use by the Horizon BCBSNJ professional staff in making coverage determinations. Members referring to this policy should discuss it with their treating physician, and should refer to their specific benefit plan for the terms, conditions, limitations and exclusions of their coverage.

The Horizon BCBSNJ Medical Policy Manual is proprietary. It is to be used only as authorized by Horizon BCBSNJ and its affiliates. The contents of this Medical Policy are not to be copied, reproduced or circulated to other parties without the express written consent of Horizon BCBSNJ. The contents of this Medical Policy may be updated or changed without notice, unless otherwise required by law and/or regulation. However, benefit determinations are made in the context of medical policies existing at the time of the decision and are not subject to later revision as the result of a change in medical policy

____________________________________________________________________________________________________________________________