SkinTE and the FDA's 361 Pathway

Doctors who perform wound treatments have limited options for skin repair. A new entrant into the wound treatment space is SkinTE, a product sold by PolarityTE. SkinTE has already been used to treat a first group of patients and has received early plaudits. The company describes SkinTE as a patient-specific, single application product created from a small piece of the patient’s own skin. As described in PolarityTE’s 2017 10K SEC filing, SkinTE is available for:

the repair, reconstruction, replacement and regeneration of skin (i.e., homologous uses) for patients who have suffered from wounds, burns or injuries that require skin coverage over both small and large areas of the body.
— from 2017 Form 10K

SkinTE is described as an impressive, all-encompassing product. The technology behind this product appears to be the company’s “TE” platform based on:

… our ability to create minimally polarized functional units, or MPFUs, which contain polarizing multi-cellular aggregates capable of expanding, proliferating and synthesizing those cells, materials, factors and/or systems we believe are necessary for integrative full-thickness three-dimensional tissue regeneration, not simply two-dimensional cell sheets. Instead of starting with artificial materials, synthetic factors and/or altered cell suspensions, our platform begins with the patient’s own (autologous) tissue and those components, appendages and substrates we believe are necessary for the development of an expandable and self-propagating complete system.
— From 2017 Form 10K

More specifically to SkinTE, the company describes the procedure and strategy:

With SkinTE, often within 24-48 hours of the initial skin harvest, our product is applied to the patient, whose body then provides a receptive environment and nutrients for controlled healing. By both preserving the tissue’s natural microenvironment and using the patient’s body as an intrinsic bioreactor—which means using the body’s own natural biological healing process rather than a manufactured or engineered environment to support the regenerative process—we believe a patient’s own tissue can be regenerated, along with its natural coloring and texture, layers and structure, hair and appendages.
— From 2017 Form 10K

This description is accompanied with a graphic overview of the SkinTE procedure that, admittedly, appears very simple to perform (right).

The company asserts that the TE platform has potential to transform tissue regeneration in multiple tissue types such as bone, muscle, fat, cartilage, nerves, and blood vessels in addition to skin. Given these strong claims, the technology behind SkinTE is certainly of interest. However, finding a detailed protocol for the technology and preparation behind SkinTE is not a trivial task.

Fortunately, Dr. Denver Lough, Chairman of the Board, President, Chief Executive Officer, and Chief Scientific Officer of PolarityTE has filed patent applications numbered 14/954,335 and 15/650,656 both titled “Methods for development and use of minimally polarized function cell micro-aggregate units in tissue applications using LGR4, LGR5 and LGR6 expressing epithelial stem cells.” This invention is largely summarized as:

[0014] The invention provides in a first embodiment a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber.
[0015] The invention provides in a further embodiment to the previous embodiment a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber where the LGR expressing cells are supplemented with growth factors and where the LGR expressing cells are selected from the group consisting of LGR4, LGR5 and LGR6.
[0016] The invention provides in a further embodiment to any of the previous embodiments a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber where the LGR expressing cells are supplemented with migratory/recruiting analytes and the LGR expressing cells being selected from the group consisting of LGR4, LGR5 and LGR6.

In effect, the invention appears to focus on a special population of LGR4, LGR5 and LGR6 expressing cells potentially supported by a matrix and supplemented with growth factors. Data behind these claims is contained in two papers:

Lough et al, 2013 Plast Reconstr Surg 132: 1159-1171 (link)
Lough et al, 2016 Plast Reconstr Surg 137: 495-507 (link)

This can be verified by side-to-side comparisons of sample figures in the above two papers and figures presented in the patent application. For example, in the first comparison below, Figures 12A and 12B from the patent application are the data published in Figure 6 of Lough et al (2013). In the second comparison, the data in Figures 5A-D of the patent application are the data published in Figure 5 of Lough et al (2016).

Patent Application 14/954,335 Figures 12A, B

Patent Application 14/954,335 Figures 5A-D

Lough et al (2013) Figure 6

Lough et al (2016) Figure 5

In Lough et al (2016), the isolation procedure for the cell population is described as generally following the protocol of Snippert et al (2010 Cell 143: 134-144), specifically maintained in:

Dulbecco’s Modified Eagle Medium/F12 (Sigma Chemical Co., St. Louis, Mo.), 10% fetal bovine serum (Atlas Biologicals, Fort Collins, Colo.), 10 ng/ml epidermal growth factor (Invitrogen, Waltham, Mass.), 20 ng/ml basic fibroblast growth factor (Invitrogen), 20 nM hydrocortisone (Sigma), 5 μg/ml insulin (Sigma), and 100 U/ml penicillin/streptomycin (Hyclone Laboratories, Logan, Utah) at 37°C in 5% carbon dioxide until passage 1.

The cells were validated for LGR6 mRNA expression, and further cultured as described prior to testing in preclinical wound models:

The scaffold/matrices were cut into 1-mm^2 pieces and transferred aseptically to 12 × 75-mm polystyrene tubes (BD Falcon, Franklin Lakes, N.J.) and 500 μl of the cells was added (1 × 10^6 total cells). The caps on the tubes were loosened to allow for gas exchange and were placed in a 37°C, 5% carbon dioxide incubator for 3 hours. The matrices were agitated every 15 minutes by gentle shaking of the tube to resuspend any cells that may have settled out. After 3 hours, the matrices were removed aseptically from the tubes and placed individually into wells of a 24-well plate containing growth media. After 24 hours, the matrices were examined under fluorescent microscopy for the adherence of the cells to the matrices. Culture medium was changed every 3 days.

In short, the procedure described by Lough et al (2016) calls for isolation of an LRG6 expressing epithelial cell population that is then cultured in a growth hormone containing medium and allowed to adhere to a matrix prior to placement in the murine wound model. As a result, the authors conclude:

In this article, we present a new method of providing skin to those wound beds that are intrinsically incapable of healing themselves.

FDA Registration Pathway

Typically, new treatments for use in the clinic must go through the FDA and be assessed for their safety and efficacy in clinical trials. However, pathways do exist to market products without safety or efficacy data in humans. One such pathway is to register with the FDA as a 361 HCT/P (Human Cells, Tissues, and Cellular And Tissue-Based Products) designation as described in Section 361 of the Public Health Service Act and 21 CFR Part 1271 (link to pdf). This is indeed the pathway used by PolarityTE. In the 2017 10K filing, PolarityTE describes SkinTE as a registered 361 HCT/P with the FDA:


This can further be confirmed via the FDA’s Human Cell and Tissue Establishment Registration portal wherein SkinTE is confirmed as an HCT/P under 21 CFR 1271.10 (screenshot right):

 

 

 

As described by the FDA, an HCT/P is regulated solely under section 361 of the PHS Act and 21 CFR Part 1271 if it meets all of the following criteria (21 CFR 1271.10(a)):

 1) The HCT/P is minimally manipulated;

 2) The HCT/P is intended for homologous use only, as reflected by the labeling, advertising, or other indications of the manufacturer’s objective intent;

 3) The manufacture of the HCT/P does not involve the combination of the cells or tissues with another article, except for water, crystalloids, or a sterilizing, preserving, or storage agent, provided that the addition of water, crystalloids, or the sterilizing, preserving, or storage agent     does not raise new clinical safety concerns with respect to the HCT/P; and

4) Either:

  1. The HCT/P does not have a systemic effect and is not dependent upon the metabolic activity of living cells for its primary function; or

  2. The HCT/P has a systemic effect or is dependent upon the metabolic activity of living cells for its primary function, and a) Is for autologous use; b) Is for allogeneic use in a first-degree or second-degree blood relative; or c) Is for reproductive use.

   
The descriptions in the patent application and published papers regarding PolarityTE’s technology raise the question of how SkinTE meets the first, second, and third criteria listed above.


Minimally Manipulated

Under the FDA guidance, skin is considered a structural tissue and serves to “physically support or serve as a barrier or conduit, or connect, cover, or cushion in the donor.” This refines the minimally manipulated criterion to ask if the processing of the tissue “alters an original relevant characteristic of the tissue relating to the tissue’s utility for reconstruction, repair, or replacement as structural tissue.”

As noted in a previous PolarityTE presentation and shown in their overview of the procedure, the SkinTE product starts as a piece of excised, full thickness skin and is then processed to yield an applied paste. This processing would appear to exceed the description of minimally manipulated, and an analogous method is specifically described as such in the FDA guidance under Example 10-4b in 21 CFR 1271.10 (right; pdf link)

Further to this point, Example 11-3 in the FDA guidance makes further clarification (right). As described in PolarityTE’s presentations and in the relevant literature, the procedure behind SkinTE no longer allows the product to maintain the “original relevant characteristics of skin related to its utility as a protective covering.” It is no longer a protective covering but rather a paste due to the processing, nor does the product retain its “dense, strong, and flexible connective tissue layer.”


Homologous Use

The FDA also mandates that products registered under section 361 meet the definition of homologous use. This generally requires the product to adequately perform its same basic function:

Under this guideline, SkinTE would have to be a product wherein its “basic” function to regenerate full thickness skin in the recipient is obvious and commonly understood, and should not require any laboratory, preclinical or clinical studies. This does not apply to the SkinTE product. In effect, if the biological function of SkinTE to regenerate full thickness skin (footnote 1) was well understood and did not require any preclinical or clinical examination, scientific papers attempting to explain and demonstrate this effect would be unnecessary. In effect, in the publication by Lough et al (2016) the authors clearly acknowledge that the group “seeks to determine” whether the LGR6 cell population isolated by fluorescent-activated cell sorting is effective for translational use (caption right). If the function of SkinTE were “basic” as described under FDA guidelines, such experiments to clarify the function of this cell population would be unnecessary.


As it stands, PolarityTE’s description that SkinTE “regenerates full-thickness hair-bearing skin” is a claim quite beyond a basic function under homologous use but rather a putatively significant treatment advancement in need of clinical validation.


Combination of Another Article

The FDA further notes that products registered under 361 may not involve “the combination of the cells or tissues with another article, except for water, crystalloids, or a sterilizing, preserving, or storage agent, provided that the addition of water, crystalloids, or the sterilizing, preserving, or storage agent does not raise new clinical safety concerns with respect to the HCT/P.” Under this guidance, products are not allowed to be mixed with a carrier during manufacture, nor are they allowed to be treated with supplements such as growth hormones during manufacture.

As shown below, both previously referenced publications by Lough in 2013 and 2016 clearly note that the fundamental research behind the PolarityTE patents involves culturing with fetal bovine serum (which contains a collection of growth factors) as well as endothelial growth factor and basic fibroblast growth factor. The conditions also call for the addition of insulin and hydrocortisone, two additional factors that would lie outside of the FDA’s guidance.

Lough et al (2013)

Lough et al (2016)

Lough et al (2016)

It’s important to note that these additives  are not limited to growth factors, nor is citation of their use confined to the published literature. For example, Lough et al (2016; caption right) as well as the cited patent filing (excerpt below) mention the use of a scaffold to support the cells in addition to the aforementioned growth factors.

 

In fact, relevant excerpts from a PolarityTE patent filing run counter to the FDA’s guidance:

  • [0014] The invention provides in a first embodiment a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber.

  • [0015] The invention provides in a further embodiment to the previous embodiment a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber where the LGR expressing cells are supplemented with growth factors and where the LGR expressing cells are selected from the group consisting of LGR4, LGR5 and LGR6.

  • [0016] The invention provides in a further embodiment to any of the previous embodiments a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber where the LGR expressing cells are supplemented with migratory/recruiting analytes and the LGR expressing cells being selected from the group consisting of LGR4, LGR5 and LGR6.

  • [0017] The invention provides in a further embodiment to any of the previous embodiments a minimally polarized micro-aggregate multi-cellular composition including isolated living LGR expressing cells and a multi-dimensional support selected from the group consisting of scaffolding, collagen, matrix, particle, and fiber where the LGR expressing cells are supplemented with LGR specific binding elements selected from the group consisting of ligand families, R-spondin, EDGF, PDGF, Wnt, VEGF, and antimicrobial peptides and where the LGR expressing cells are selected from the group consisting of LGR4, LGR5 and LGR6.

As clearly described in the bolded segments, the patent filing by PolarityTE consists of claims which include combinations with other components such as scaffolding and growth factor receptor ligands. These additives are not allowed for products registered under the 361 pathway. It remains possible that PolarityTE intentionally removed both the scaffold and growth factors from the marketed version of SkinTE. At a recent investor conference, the company mentions the use of “crystalloids” as a carrier rather than scaffolds. But if they have removed both scaffolds and growth factors, does the product used to treat patients meet the company’s own efficacy claims as published in the scientific literature and related patent filings? What is the final SkinTE product?

 

In sum, PolarityTE’s registration of SkinTE as a 361 HCT/P has allowed the company to rapidly bring their product to market for sale and use in the clinical setting. However, the FDA guidelines regarding the standards for a 361 HCT/P designation and the SkinTE product as described in whole or in part through PolarityTE presentations, patent filings, and scientific publications authored by the CEO raises questions. Are the production methods behind SkinTE and the final product offered by PolarityTE compliant with the FDA’s 361 standards? Have the scaffolds and growth factors described in the scientific publications and patent filings been omitted from SkinTE? Absent appropriate growth factors and culturing conditions, how do the cells in the excised skin maintain their differentiated state and pattern of LGR receptor expression? Are specific LGR receptor expressing cells isolated via fluorescent-activated cell sorting for use in SkinTE? Has the FDA effectively examined products like SkinTE to assure the public that the standards outlined by 21 CFR 1271 are met and adhered to? In this specific case, PolarityTE is marketing a product that has not been tested in randomized controlled clinical trials. Whether this treatment choice meets the FDA guidelines under section 361, and whether this choice is clinically safe and efficacious are questions in the public’s interest that should be further examined.

 

 


Footnote 1: PolarityTE makes a clear claim that “SkinTE Regenerates Full-Thickness Hair-Bearing Skin.” Screenshot below taken from the PolarityTE website.

 

Is Eteplirsen an Exceptional Case for the FDA?

Prospective Duchenne muscular dystrophy (DMD) treatment eteplirsen faced an uphill battle at the April 25th FDA advisory panel (replay here). Earlier release of the briefing documents had underlined the FDA’s persistent concerns about the sponsor Sarepta’s data package, leaving most observers skeptical about the panel’s chance at a positive outcome. The advisory panel itself was a long affair marked by an open public hearing full of resolute and emotional statements made by young boys with DMD, parents of boys with DMD, medical doctors and researchers. 

At the end of the ~11 hour panel, those previously unconvinced of the drug’s efficacy were likely in agreement with the negative tallies that accrued for the voting questions. Nonetheless, those hopeful for the drug’s prospects perceived two significant lifelines cast out by medical doctors as well as by prominent FDA staff members.

 

The Doctors

Medical doctors who had treated boys with eteplirsen voiced a unanimous message: boys with DMD who had received eteplirsen performed unequivocally better than other boys in their practice history. Such comments understandably bolstered the confidence of advocates and families pushing for approval. Although encouraging, the anecdotal comments by the doctors are unlikely to trump the accrued clinical data, for a few reasons:

  1. Doctors should indeed be advocates for their individual patients and no one should want a doctor who isn't. In that regard, it is not unreasonable or out of place to see individual doctors lobby on behalf their patients, even if the drug in question provides an ambiguous outcome.
  2. Clinical trial history has shown anecdotal individual experience, either by a patient or a treating physician, to be no substitute for larger controlled trials. In this case, the statements by the doctors left no room to suggest any overlap between the performance of boys on eteplirsen versus previous boys in their practice. Unfortunately, the clinical data are not indicative of such a uniformly exceptional experience.

Shown below are figures from Sarepta’s briefing documents (Fig. 19) and the FDA’s (Fig. 2).

Perhaps some advocates can reconcile the uniformly exceptional performance cited by the testifying clinicians against the clear overlap of the eteplirsen and control data sets. However, to neutral observers, such graphs are unlikely to mesh with their mental image of an unequivocal effect. It’s understandable that the doctors who participated in the open hearing are advocating for their patients, but this advocacy has little scientific backing.

 

The FDA

Ultimately, the decision of approval lies with the FDA. Adam Feuerstein’s recent article builds on this theme and assembles clues to buttress Sarepta’s case. Initially, he notes:

“If Woodcock [FDA Director, Center for Drug Evaluation and Research] agreed with all the bad things said about eteplirsen on Monday night, if she had no plans to overrule or cast aside the recommendations against approval, why did she go out of her way to meet with DMD boys and their families immediately after the meeting ended?
Could the answer be that Woodcock plans to approve eteplirsen?”

An observer could certainly sense positive vibes from this type of post-panel interaction, if they were so inclined. However, this meeting could just as easily be described as normal social behaviour. In fact, Monday’s panel was not the first time that Dr. Woodcock had met these families. In February of 2013, Drs. Woodcock and Robert Temple of the FDA met with the Jett Foundation, a group working to improve the lives of those with DMD. After that meeting, the group wrote:

“Our conversation with the FDA leaves us positive and optimistic. These high ranking FDA officials were extremely supportive, engaging, and receptive to our ideas about accelerated approval. It was our collective impression that the FDA seemed committed to working as expeditiously as possible to grant access to life saving treatments to those who will benefit. We were able to start the dialogue that will affect how Duchenne is viewed by the regulatory agency. We appreciate that the FDA is allowing patients, parents, and advocates to play an integral role in the regulatory process. We will continue our work with the FDA as more drugs begin to reach trial level. Our goal is that all Duchenne patients get treatment as quickly as possible.”

In that regard, Dr. Woodcock is simply being a normal, decent human being. She has met these families before and no doubt cares for their compelling cause. There is little reason to believe she would have ignored these parents even if the agency was on track to reject the application for marketing. To further buttress this interpretation, we can go back to 2013 once again. Despite the positive interaction the Jett Foundation had with Drs. Woodcock and Temple in February, November 2013 saw the FDA push back a potential filingby eteplirsen’s sponsor.

 

Later on during the panel, a theme emerged regarding the interpretation of the presented data and testimony: are we to believe the graphs and the underlying statistics, or should we give strong weight to the doctor and DMD community testimonies?

Dr. Woodcock explains:

“The standard is adequate and well controlled trials, OK, that's what is in this statute, but we are instructed to have flexibility on how we interpret that based on medical need.”
“We are instructed, as people said, to take the views of the patient community into account, more on the benefit and the risk. So, the statutory standard is more or less as described there but there is flexibility and that's where we should take the views of the community into account.”

As Feuerstein notes, the FDA’s Ellis Unger added this clarification:

“I think with the majority of the [eteplirsen] patients here, we have an incredible advantage in my time at the FDA that is unprecedented. To have all the patients here so that is an important advantage we have. One thing you can try to do is reconcile what you heard from the patients with the data that you've seen presented by the company. We're hearing patients are improving, doing things next year that they didn't do last year, and you have to figure out if you can reconcile that with the actual hard data you've been analyzing today.”

The suggestion is the above two comments tip the FDA’s position towards listening to the patients. Perhaps. But Dr. Unger is very clear the testimony and the actual data are at conflict, and his comments could simply be interpreted as a neutral acceptance the conflict is there and requires resolving by panel members. Woodcock’s comment regarding flexibility is true, but could also neutrally be viewed as a simple recitation of the standard under which the FDA operates.

Subsequently, Feuerstein outlines the following exchange between panel member Dr. Chiadi Onyike and the FDA’s Robert Temple:

Dr. Onyike: The question twice mentions 'well controlled,' and as you've heard repeatedly, some people have said they have trouble with the controls. So, this 'well controlled' phrase in a sense tips or constrains the question.
Dr. Temple: I understand a lot of people don't like historically controlled trials. They're not sure they believe they're well controlled. Our regulations since 1970 have said that a historical controlled trial can be adequately and well controlled study. The question here goes: Under the circumstances do you think it was? Do you think the way they selected patients, the way they analyzed them, was good enough to make it an adequate and well-controlled study? That's the question. Historically, historical-controlled trials have been the basis for approval -- sometimes in sort of obvious cases and sometimes in cases that quite aren't so obvious.

This is interpreted as the FDA “saying that comparing data collected from a single arm study against a historical control -- like what Sarepta did -- is definitely okay.” Absolutely. But this is again a recitation of FDA guidelines. Dr. Temple provides no hint the FDA believes Sarepta’s historically-controlled study is adequate; rather, he simply states the FDA would accept one, provided the sponsor conducted an adequate one, and that panel members should not see the use of a historical control arm to be disqualifying.

In sum, although many of the actions and statements at the panel can be weaved into a thesis for approval of eteplirsen, their simplest explanation supports a view of the FDA as fastidious and thorough in its duties. They asked panel members to reconcile data and testimony, clarified the prevailing statutes, and provided a forum for the DMD community to express their views.

 

Perhaps the Sarepta story is best summed up by Mr. Benjamin Dupree, a 23 year old with DMD who served as a patient representative on Monday’s panel. Mr. Dupree voted Yes in Vote 7, wherein panelists were asked:

“Do the clinical results of the single historically-controlled study (201/202) provide substantial evidence (ie, evidence from adequate and well-controlled studies or evidence from a single highly persuasive adequate and well-controlled study that is accompanied by independent findings that substantiate efficacy) that eteplirsen is effective for the treatment of DMD?”

When given a chance to comment on his vote, Mr. Dupree was overcome with emotion while saying the following:

“I voted yes. I can’t really reconcile the difference between the testimony that was given suggesting that boys are recovering abilities. I don’t… living with Duchenne I don’t understand how that’s even possible but at the same time the study doesn’t prove from a scientific… it doesn’t provide what i think is adequate evidence to support all this testimony that I am seeing in here.”

It’s not useful or fair to question how Mr. Dupree reconciled his Yes vote with his admitted difficulty in seeing adequate scientific evidence. That he was so emotionally overcome hints, at the least, to a part of Mr. Dupree that wishes the scientific data behind eteplirsen were as strong as the efforts of the DMD community.