Monday, August 29, 2011

Does FDA’s expanded access program serve the best interests of terminally ill patients?

Background
In 2007 FDA once again broadened its policy for expanded access to new drugs when it amended the Food and Drug Administration Modernization Act (FDAMA) to include criteria for approved drugs with risk evaluation and mitigation strategies (REMS).(Fed Reg 2009;74(155)) This marks the 3rd major revision in 3 decades to regulations that grant seriously ill patients increasingly open access to investigational drugs.

Access to investigational drugs via treatment Investigational New Drug (IND) applications was first formalized in 1987 following intense pressure from AIDS activists. Supporters applaud each move towards easier access to unapproved drugs, but the regulation, in essence, reverses elements of the Food, Drug, and Cosmetic Act (FD&C) which were put in place to protect all patients from unsafe medications and charlatanism. This nullifies the attempts to protect the best interests of patients, including those who are terminally ill.

In the context of this paper I define “best interests” as: meeting one’s fundamental rights to self-preservation, ensuring one’s safety, ensuring no person or persons has fewer or more rights than others, and ensuring benefits are equitably distributed. This definition corresponds to the criteria listed in the “Rule” below.



ISSUE:
Does FDA’s expanded access program serve the best interests of terminally ill patients?


RULE:

FDA’s expanded access program serves the best interests of terminally ill patients if all of the following are true:
  1. Expanded access meets a fundamental right of access to investigational drugs
  2. The benefits of using an investigational drug outweigh the risks and the patient is not exposed to unreasonable and significant additional risk of illness or injury, even if the patient is already terminally ill.
  3. Terminally ill patients do not need the same protection against unsafe or ineffective drugs as patients with less serious or non-life-threatening illness.
  4. All terminally ill patients have equal access to the drugs in question.
  5. Expanded access does not interfere with development of new drugs and thereby negatively affect future access to other patients.


ANALYSIS:

CRITERIUM 1: Expanded access meets a fundamental right to access investigational drugs

The current laws do not support fundamental right to access investigational drugs. According to the FD&C Act, “no person shall introduce or deliver for introduction into interstate commerce any new drug” unless it has been approved for marketing by FDA. Approval requires that the new drug has been proven both safe and effective in humans based on at least 3 phases of “adequate and well-controlled” clinical studies. [See 21 CFR 312.21 Phases of an investigation and 21 USC 355 Section (d)] Drug sponsors are permitted to submit treatment INDs to provide Phase 3 or, less commonly, Phase 2 drugs to patients with “a serious or immediately life-threatening disease condition in patients for whom no comparable or satisfactory alternative drug or other therapy is available.” [See 21 CFR 312.34] However, FDA reserves the right to deny access if there is “insufficient evidence of safety and effectiveness to support such use” or there is an “unreasonably and significant additional risk of illness or injury,” even if the patient’s illness is immediately life-threatening. [See 21 CFR 312.34] Furthermore, no one can compel the drug sponsor to provide investigational drugs to patients not enrolled in clinical studies, or make such a request on their behalf; participation in a treatment IND is voluntary. So the current laws do not support the existence of a fundamental right.

This argument is supported by the 2007 ruling in a recent case: Abigail Alliance for Better Access to Developmental Drugs v. von Eschenbach. The Abigail Alliance was formed by Frank Burroughs after his daughter, Abigail, died of head and neck cancer in 2001. She had been denied access to then-investigational treatments cetuximab (Erbitux) and gefitinib (Iressa), both of which went on to win FDA approval after her death. Convinced the denial of access (and possibly Abigail’s premature death) was caused by unnecessary regulation and bureaucracy, the Abigail Alliance sued FDA (Andrew von Eschenbach was Commissioner at the time) in an attempt to change the existing regulations and permit terminally ill patients to access new drugs as soon as they have completed Phase 1 trials.

In its argument the Abigail Alliance claimed “the concepts of self-defense, necessity, and interference with rescue are broad enough to demonstrate the existence of the fundamental right they seek—a right for ‘persons in mortal peril’ to ‘try to save their own lives, even if the chosen means would otherwise be illegal or involve enormous risks.’” In support of its argument, the Alliance cited Washington v Glucksberg saying FDA’s regulations violated the Constitutional concept of Due Process and the right to self-protection. But the US Court of Appeals for the District of Columbia dismissed the argument by saying, “The Alliance’s effort to focus on efficacy regulation ignores one simple fact: it is unlawful for the Alliance to procure experimental drugs not only because they have not been proven effective, but because they have not been proven safe.” The Court disagreed with the Abigail Alliance’s interpretation of Washington v Glucksberg stating, “A tradition of protection does not alone establish a fundamental right."

In addition, the US Court of Appeals for the District of Columbia asserted, “There is no fundamental right ‘deeply rooted in this nation's history and tradition’ of access to experimental drugs for the terminally ill.” The Abigail Alliance took its appeal to the US Supreme Court, but they were denied certiorari in 2008 and the District Court decision stood.

I feel it is also important to add here that a fundamental right should not, and could not, apply to only to a segment of the population (terminally ill people). If the right is truly fundamental, all persons should have the same access to investigational drugs even if their illness is mild and non-life-threatening. Current laws and the Constitution include no such fundamental right, and so this first criterium is not met.


CRITERIUM 2: The benefits of taking investigational drugs outweigh the risks

The FD&C Act was enacted in 1938 after 100 Americans died as a result of taking an unregulated formulation of elixir sulfanilamide. The drug had been given successfully in tablet form to treat streptococcal infections, but when it was dissolved in diethylene glycol to create a liquid form the results were lethal. FD&C therefore required sponsors to demonstrate the safety of their drugs before being able to sell them. The subsequent 1962 Kefauver-Harris Amendments to FD&C required sponsors to demonstrate that new drugs were also effective by completing 3 phases of clinical trials and gaining approval on a New Drug Application (NDA). The Amendments were meant to protect patients against charlatans who made false promises to cure ill people, including patients with terminal diseases.

Phase 1 clinical trials are conducted in a handful of healthy volunteers to “determine the metabolism and pharmacologic actions of the drug in humans, the side effects associated with increasing doses.” Phase 2 clinical trials are conducted in no more than a few hundred patients with the disease under study to “determine the common short-term side effects and risks associated with the drug.” It isn’t until Phase 3 that trials are conducted in a wider population (several hundred to several thousand patients with the disease under study) that the sponsor is able to “gather the additional information about effectiveness and safety that is needed to evaluate the overall benefit-risk relationship of the drug and to provide an adequate basis for physician labeling.” [See 21 CFR 312.21]

The expanded access program violates the protection afforded by FD&C by exposing patients to drugs that do not have proven safety or efficacy profiles. Medical and physiological responses of healthy Phase 1 volunteers are not representative of the effects of a drug in sick patients who may suffer from concomitant illnesses, have compromised abilities to metabolize drugs or manage side effects, and may be taking other medications. Even Phase 2 and 3 trials may not be representative of the larger population afflicted with a given disease because the number of trial participants is small compared to the total population, and the controlled conditions of clinical trials differ from the “real world” experience of patients.

Furthermore, only 11% of drugs that enter clinical trials are ever approved for marketing. For cancer drugs the approval rate is 6%. (Okie 2006, Society for Clinical Trials Board of Directors 2006) Researchers have been surprised in the past when drugs that appeared to be effective and safe in Phase 1 or 2 trials turned out to be ineffective, dangerous or both once the drug was used to treat a larger population in Phase 3. [Society for Clinical Trials Board of Directors 2006, Cortez 2011, Ray 2011] Of course, some drugs are recalled or have additional warnings added to their labels after they are approved for marketing. [eg, Vioxx, Meridia, Avastin, Avandia, Darvon & Darvocet, Depo-Provera, Plavix] This demonstrates the limitations of even Phase 3 trials in establishing safety and efficacy in the broader population.

Investigational drugs have, by definition, an incomplete safety and efficacy profile. So the expanded access program cannot predict whether the benefits of taking investigational drugs outweigh the risks. Therefore this second criterium is not met.


CRITERIUM 3: Seriously or terminally ill patients do not need the same protections as people with less serious disease

There is no legal precedent indicating seriously ill people, even those with terminal disease, have less need for protection than people with less serious diseases. In United States v Rutherford cancer patients argued that they had a right to Laetrile, an investigational drug that was said to be useful in treating cancer. Laetrile was never shown to be effective and was never approved by FDA, but in their arguments the plaintiffs claimed that denying them access was an infringement on their Constitutional privacy rights to benefit their personal health. They also claimed that the safety and efficacy standards outlined in FD&C are not reasonable applicable to terminally ill cancer patients.

A District Court sided with the plaintiffs and a Court of Appeals approved the District Court’s decision, but on certiorari the US Supreme Court reversed the decision. In the unanimous opinion Justice Thurgood Marshall wrote: “Nothing in the history of the 1938 Food, Drug, and Cosmetic Act, which first established procedures for review of drug safety, or of the 1962 Amendments, which added the current safety and effectiveness standards in § 201(p)(1) suggests that Congress intended protection only for persons suffering from curable diseases. To the contrary, in deliberations preceding the 1938 Act, Congress expressed concern that individuals with fatal illnesses, such as cancer, should be shielded from fraudulent cures...For the terminally ill, as for anyone else, a drug is unsafe if its potential for inflicting death or physical injury is not offset by the possibility of therapeutic benefit.”

I would also add that terminally ill people are more at risk for being manipulated because they are making decisions under duress. They are more likely to believe a treatment will be helpful because it provides them with hope, whereas if their illnesses were not life-threatening they may be more wary and skeptical of the same investigational drugs.

Just as the Declaration of Helsinki protects vulnerable populations (prisoners, children, pregnant women) those who are desperately ill need protection, even if they don’t always perceive regulations to be protective. The expanded access program fails to protect terminally ill people in the same way as people with non-life-threatening diseases, because it allows them to be exposed to unknown risks that are otherwise considered unacceptable for the rest of the population.

The program also falsely raises the hopes of the terminally ill simply because by granting early access FDA implies the drugs are acceptable for use. Due to these failures of protection, the third criterium is not met.


CRITERIUM 4: All terminally ill patients have equal access to the drugs in question


Expanded access was meant to make treatments available to terminally ill patients who do not qualify for clinical trials and who have no alternative approved treatments. However, this access is not equal.

First, to apply for the drug an individual patient (as opposed to a population represented by a drug sponsor) needs a physician who is dedicated and willing to complete the treatment IND. Not all treating physicians are willing or able to commit their time to this work and some physicians may be uneducated on the process or even the availability of a treatment IND. So a patient may be denied access based on their choice of physician (if they had a choice).

Second, sponsors are permitted to charge for investigational drugs so long as they are only recouping their costs to manufacture the drug. Insurance companies generally do not cover investigational drugs so the cost, which can be considerable, may fall to the patient. This favors patients with more financial resources. (Sponsors can choose to waive costs, but they are not required to do so.)

Third, even if drug sponsors are willing to take on treatment INDs their eligibility criteria can exclude some patients. Drug sponsors want to get the best results possible and so they are prone to recruiting patients who are most likely to respond well. This bias will always prevent some portion of the population from equal access to investigational drugs.

Fourth, investigational drugs are typically manufactured in small quantities. Even if the sponsor is willing to provide the treatment free of charge, there simply may not be enough medication available for all patients who apply for it through the expanded access program. And the sponsor cannot be compelled to make enough of the drug to meet all possible demand.

Fifth, patients can be disadvantaged by their geographical location. If a condition of treatment is proximity to a location where the drug can be administered, patients who are unable to travel (for financial, health, or any other reasons) are excluded.

For these reasons the fourth criterium is not met.


CRITERIUM 5: Expanded access does not interfere with development of new drugs (and therefore decrease access to the wider population of patients)

Drug sponsors have legitimate reasons not to want to participate in expanded access. For example, expanded access can decrease enrollment in clinical trials. Patients who are terminally ill generally do not want to risk being randomized to placebo or existing treatment arm if they think the investigational drug offers more hope. So they opt out of the clinical trial and apply instead for expanded access. Each time FDA loosens restrictions around investigational drugs the demand for access outside of clinical trials grows. This makes it more difficult for sponsors to recruit participants for controlled studies.

Additionally, unfavorable side effects, injuries, or deaths resulting from the use of investigational drugs outside of clinical trials results must be reported to FDA and can put post-approval marketing efforts at risk. Sponsors are understandably skittish about potential negative impacts on revenue.

Finally, expanded access exposes drug sponsors to potential lawsuits. In several recent cases patients have also sued sponsors for not making investigational drugs available to them indefinitely. [See Abney v Amgen and Suthers v Amgen] This legal and financial exposure is a deterrent to sponsors.

The potential for negative impact on clinical trials and safety data threatens profitability and can drive sponsors to curb research and development (R&D) investments in new drugs that are targets for expanded access (eg, serious but relatively uncommon diseases). This interferes with the development of new treatments and can unfairly prevent future patients from access to life-saving drugs.

For these reasons, the fifth criterium is not met.


CONCLUSION:
While terminally ill patients can sometimes benefit from getting investigational treatments, the expanded access program does not serve the best interests of terminally ill patients.



Monday, August 8, 2011

Guidance for Industry: Enforcement Policy OTC Sunscreen Drug Products Marketed Without an Approved Application


Name of Guidance:


Guidance for Industry: Enforcement Policy OTC Sunscreen Drug Products Marketed Without an Approved Application


Status of Guidance:


Draft guidance, final expected to be issued in 2011


Date of Guidance:

 June 2011


Names of Organizations Releasing the Guidance:


United States Department of Health and Human Services (HHS)
United States Food and Drug Administration (FDA)
Center for Drug Evaluation and Research (CDER)

Target Audience:


Manufacturers of over-the-counter (OTC) sunscreen drug products without an approved application.

Guidance Laws and Regulations Referenced in the Guidance:


21CFR 201.327, §201. §327(c)(3), §201.327(g), §201.327(l): FDA guidances pertaining to SPF labeling and testing requirements for OTC sunscreens.
21 CFR 352.10, 21CFR 352.20
21CFR 330.1: Guidance addressing GRASE drugs, ie, drugs generally recognized as safe and effective.
21CFR 310.545(a)(29)(ii): Guidance regarding active ingredients found in OTC drugs, including sunscreens.




Definitions:

Drug monograph: A description of a drug or class of drug that specifies their ingredients, directions for use, conditions of use, and contraindications.

GRASE: Acronym for Generally Recognized as Safe and Effective.

OTC: Over-the-counter. Here, OTC sunscreens are those sold directly to customers without a prescription.

SPF: Sun protection factor. A rating; for sunscreens from 2 to 50 plus, that indicates the extent of protection provided by a sunscreen.

UVA and UVB: Ultra-violet rays A and B, which are responsible for tanning, but also for sunburn, skin damage, or sometimes skin cancer.


Background:


Some over-the-counter (OTC)  sunscreens appear on the market without FDA application approval. Since there is no final description, or drug monograph, of this class of drug, the FDA has been unable to finalize guidances pertaining to these products. Concern over safety and effectiveness led the FDA to issue numerous publications to guide manufacturers by expressing its viewpoint. This draft guidance is the most recent publication on the topic and the final guidance, pending the drug monograph and other studies, should also be available in 2011. The main concerns regarding OTC sunscreens are testing and labeling requirements and proposed enforcement policy.

Summary:


Numerous Federal Register notices have be published by the FDA on the topic of OTC sunscreens. Key dates follow:

1978: Publication of notice of intent to address safety issues (SPF and GRASE).

1993: Safety issues addressed. A minimum SPF value of 2, as well as views on safe dosage forms are proposed.

1996-2000: Several active ingredients added to list of those eligible for inclusion in drug monograph.

2007: Safety and effectiveness discussed. Products containing insect repellent examined.

2011; Labeling and testing requirements modified (21CFR 201.327) for marketed sunscreens without application approval. SPF label upper limit of 50 plus required. Dosage forms eligible for inclusion in drug definition proposed.


Because a drug monograph has not yet been finalized for .OTC sunscreens, the FDA intends to use enforcement discretion until the monograph is finalized. Additional safety and efficacy data is being collected and will be incorporated in the final guidance expected to be issued in 2011. Other issues to be addressed in the final guidance include active ingredients, special warnings on sprays, and combination sunscreen and insect repellent.


Rationale:

 Manufacturers are currently marketing sunscreens without approved applications since there are no final requirements for testing and labeling of these products at this time. In addition, a final drug monograph of sunscreens is not yet completed. This guidance expresses the FDA’s expected finalized recommendations and intended enforcement approach. Manufaturers of OTC sunscreens should follow these recommendations while the final guidance is pending.

 Resulting Recommendations:


The following recommendations express the FDA’s intended enforcement policy pending finalization:

·        OTC sunscreens may be marketed without application approval if they are not potentially dangerous to use.

General safety issues concern:

o   Safety of active ingredients (21 CFR 352.10 and 352.20)

o   Appropriate claims in labeling (21CFR 201.327(c)(3) and (g) and 310.545(a)(29)(ii)

o   Adverse event reporting (21CFR 330.1) and adulteration.

o   Labeling and testing requirements (CFR 201.327)

·        In vitro broad spectrum testing must be done to test for protection across different UVA wavelengths (21CFR 201.327(i)).

·        OTC sunscreens claiming to have an SPF of over 50 should be marked 50+ or 50 plus,

·        Dosage forms differ in eligibility for inclusion in future drug definitions. Sprays.

should be labeled with special warnings to prevent inhalation, in compliance with 21CFR 201.327(e)(l)(ii).


Impact:

 This draft guidance serves to inform manufacturers of OTC sunscreens without approved applications of the FDA’s intended enforcement approach in regards to testing and labeling of these products. This will facilitate adherence to the final FDA guidance, expected to be issued in 2011.

Sunday, August 7, 2011

Brief Summary: E15 Definitions for Genomic Biomarkers, Pharmacogenomics, Pharmacogenetics, Genomic Data and Sample Coding Categories

Name of Guidance

E15 Definitions for Genomic Biomarkers, Pharmacogenomics, Pharmacogenetics, Genomic Data and Sample Coding Categories

Status of Guidance

Final Guidance

Date of Guidance

April 2008

Released by

Center for Drugs and Biologics, Federal Drug Administration, Department of Health and Human Services

Link to Guidance

Target audience

Developers of drugs/biologics

Laws and Regulations

This guidance does not refer to any laws or regulations.

Definitions

Biomarker: Any substance used to detect and identify a type of cell, organelle, sub-component, biological state, or biological process

Deoxyribonucleic acid (DNA): genetic material that comprises an organism

Genome: the ordering of an organism’s full DNA sequence (or genetic code)

Genomics: the branch of genetics that focuses on organisms in terms of their genomes (or DNA sequences)

International Conference on Harmonisation (ICH): With the full name of the International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use, the ICH is a global organization with the mission of creating a single procedural platform for regulatory and pharmaceutical entities worldwide, specifically in Europe, Japan and the US, to follow to “harmonize” the steps involved in drug development and registration.

Pharmacodynamics (PD): the study of the biochemical and physiological effects of drugs, and the mechanisms of their actions, including the relationship of their actions and effects with their chemical structure

Pharmacokinetics (PK): the area of pharmacology focusing the effect of body processes on drugs, including absorption, distribution, metabolism, and elimination

Ribonucleic acid (RNA): a nucleic acid composed of repeating nucleotide units of ribose sugar, phosphate group, and nitrogenous base that is involved in protein synthesis. RNA is different from DNA in that it includes the sugar ribose and the base uracil while DNA includes the sugar deoxyribose and the base thymine.

Single nucleotide polymorphisms (SNPs): a deviation in a sequence of DNA that involves the switch of a single nucleotide base for its alternate, as in replacing the cytosine base with the thymine base. These variations occur at an increasing rate in the population.

Background

The International Conference on Harmonisation (ICH) formed to promote the use of the same procedures and documents in the development and registration of drugs by regulatory and pharmaceutical entities worldwide. Formerly, each region of the world followed its own procedures. Part of the problem with having different procedures around the world is the accompanying discrepancy on the meanings of vocabulary used in these processes. In an effort to clarify and consolidate the vocabulary and to streamline communication, the ICH advocated for the adoption of specific vocabulary to be used with agreed-upon definitions.

Another area in drug development that needed consolidation and clarification is the labeling of biological samples that are used for research in the fields of pharmacogenomics and pharmacogenetics. Different countries use different systems for identifying and tracking these samples and the data collected on these samples. Accordingly, the ICH called for a single system for coding for these samples and data that would simplify research and the development of new drugs.

Summary

This guidance is endorsed by the ICH and provides agreed-upon definitions for commonly used words in the fields of pharmacogenetics and pharmacogenomics, and in genomic data and sample coding.

Rationale

Lack of consistency of definitions for commonly used terms in drug development around the world creates great confusion, creates work, and wastes money. Creating a system in which all parties involved in drug development and registration around the world can communicate clearly would address and end this confusion and waste, and ensure proper interpretation and communication while making drug discovery more efficient.

Resulting Recommendations

This guidance provides the following definitions to clarify meanings of terms and to ensure consistent understanding.

· Genomic biomarker: a measurable DNA and/or RNA characteristic that is an indicator of normal biologic processes, development of disease, and/or response to therapy (eg, the expression, function, or regulation of a gene)

· Pharmacogenomics (PGx): the study of variations in the characteristics of DNA and RNA as related to drug response in terms of drug discovery, drug development, and clinical practice

· Pharmacogenetics (PGt): a subset of pharmacogenomics, focusing on the study of variations in DNA sequence as related to drug response drug discovery, drug development, and clinical practice

· Drug: refers to investigational (medicinal) product, medicinal product, medicine, and pharmaceutical product (including vaccines and other biological products)

· Drug response: the processes of drug absorption and disposition (meaning pharmacokinetics), and drug effects (meaning pharmacodynamics, drug efficacy, and adverse effects of drugs).

Samples, and the data generated by them, can be labeled with one or two codes during research. This guidance recommends using a system including the following 4 specific categories for coding biological samples (and the generated genomic data).

· Identified: the samples and/or data are identified with personal information to allow tracing back to the subject from whom the sample/data was derived. Samples/data with this code can be withdrawn or returned, permitting clinical monitoring, subject follow-up, and future addition of new data from the identified subject. Clinical trials in drug development typically do not use identified samples/data.

· Coded: these samples/data do not include any personal information and are tagged with one (single-coded) or two (double-coded) codes. Single- and double-coded samples/data can be traced back to the subject, so sample withdrawal and return of results are possible. It also enables clinical monitoring, subject follow-up, and future addition of new data from the identified subject. Single-coding is the most commonly used method in clinical research today. Double-coding allows an extra layer of security on samples/data.


· Anonymized: these data/samples are single- or double-coded and increase the level of confidentiality and privacy because they do not include personal information on the subject from whom the sample came. The samples/data are not traceable back to the subject so it is not possible to withdraw the sample, return the results, conduct clinical monitoring or subject follow-up, or add new data from the subject at a future date.


· Anonymous: these samples/data are completely anonymous, with no personal information included, voiding any possibility to trace data back to the subject. The samples/data are not traceable back to the subject so it is not possible to withdraw the sample, return the results, conduct clinical monitoring or subject follow-up, or add new data from the subject at a future date.


Impact

Using an agreed-upon set of vocabulary and coding categories for biological samples (and their genomic data) will improve communication and streamline drug research worldwide.

Monday, August 1, 2011

Guidance Document for the Preparation of IDEs for Spinal Systems

Status of Guidance:
Final

Name of Organization:
U.S. Food and Drug Administration
Center for Devices and Radiological Health

Date of Guidance:
January 2000

Target Audience:
Makers of spinal devices who wish to apply to the U.S. Food and Drug Administration (FDA) for an Investigational Device Exemption (IDE)

Laws and Regulations Referenced:
21 CFR 25: Describes the FDA’s considerations relating to environmental impact
21 CFR 50: Describes how human subjects are to be protected in clinical trials
21 CFR 56: Describes the role of institutional review boards
21 CFR 58: Describes good laboratory practices studies that are not conducted on humans
21 CFR 801: Describes the labeling requirements for medical devices
21 CFR 812: Outlines the requirements of an IDE application
21 CFR 860: Explains how device classification is determined based on safety and effectiveness data

Definitions:
Investigational Device Exemption (IDE): The FDA grants device makers the ability to test their new devices in humans by approving an IDE.

Spinal system: A complete spinal implant including all parts.

Component: An individual part of a spinal system.

Background:
The latest spinal devices are often designed in totally different ways than older devices. They may use new materials and pose greater risks along with potentially new benefits to patients. Before a new device can be tested in humans, the device maker must apply to the FDA for an IDE. The IDE includes all details of how the clinical studies will be conducted and how the results will be interpreted. This guidance outlines the FDA’s areas of concern specific to IDEs for spinal devices.

Summary:
Before testing a new spinal device in humans, the device maker must show that the new product appears to be reasonably safe. They may do this in various ways including:

(1) Citing scientific articles describing clinical trials conducted in other countries or on similar devices or components,

(2) Providing the results of animal testing to show that the material will likely be safe in the human spine, will stabilize the spine as intended, and will promote or prevent fusion, depending on the intended use,

(3) Providing the results of mechanical testing showing the device withstands certain weights (static testing) and repetitive motions (fatigue testing).

After establishing preliminary safety, the device maker describes the details of the human studies they plan to conduct. The applicant states the goals of the study and proposes a study design. The preferred design is a randomized control trial, in which one group of patients (control group) is given a standard treatment and a similar group (experimental group) is given the new treatment and the outcomes compared.

The IDE application should outline the factors that will determine whether a patient is eligible for the study. Patients may be included if they are within a specific age range, have a certain condition affecting a specific area of the spine, and are deemed likely to adhere to the follow-up schedule. Patients may be excluded if they have other diseases that could affect results, if they have had previous surgeries, are pregnant, or are deemed unlikely to follow up.

The number of patients to be included in the study should ensure that results are statistically meaningful. Patients should be followed for at least 2 years. The FDA recommends that patients be evaluated at the start of the study (before surgery), and 2 weeks, 3 months, 6 months, 1 year, and 2 years following surgery.

There are many ways to determine the success of a new spinal implant. The FDA notes that the outcomes should be both statistically and clinically meaningful. This means that there should be a measurable (objective) improvement in range of motion and/or anatomical measurements of the spine, as well as a patient-reported (subjective) improvement in pain and functionality.

Safety of the implant should be evaluated by documenting any repeat surgeries or adverse events during the follow-up period, and testing for any neurological problems that did not exist prior to surgery.

The IDE application must also describe how statistical methods will be used to analyze the study results. This explanation should include how the sample size was determined, and how trends in the data will be detected.

Finally, the IDE must include proposed labeling for the device. The package label, the information contained inside the package, and the manual describing the surgical technique for implanting the device must all clearly state that the product is an investigational device.

Rationale:
Spinal systems are complex medical devices that can pose serious risks that are unique to their design and their uses in the spine. The FDA must evaluate the safety and effectiveness of these devices in humans before approving them for widespread use. Device makers must create a lengthy and complex IDE application that addresses the FDA’s concerns specific to these devices. This guidance outlines those areas of concern to help device makers produce a complete IDE application.

Resulting Recommendations:

· Device makers should consult the FDA early in the development of proposed human testing of their devices.

· Devices should be shown likely to be safe in the human spine through evidence in the scientific literature, animal testing, and mechanical testing.

· The proposed clinical study should have a clear hypothesis and objective.

· The proposed clinical study should include enough patients to make it statistically relevant.

· Study participants should be randomly assigned to either a control group or the testing group.

· Patient inclusion and exclusion criteria should be defined in the IDE application.

· Patients should be followed up for at least 2 years.

· Effectiveness of the device should be determined by both objective measurements and reports of patient satisfaction, including improvements in pain and function.

· Safety should be evaluated by analyzing adverse events, reoperations, and neurological problems.

· Planned statistical analyses should be described in the IDE application.

· The IDE application should include proposed package labeling, informational insert, and surgeon instructions, all of which should identify the device as investigational.

Impact:
By explaining the FDA’s main areas of concern relating to the safety and efficacy of spinal devices, this guidance should shorten the time to ultimate device approval by helping device makers submit a successful IDE application on their first attempt.