Medical Device Development Tools. Draft Guidance for Industry, Tool Developers, and Food and Drug Administration Staff

Similar documents
FDA Decisions for Investigational Device Exemption (IDE) Clinical Investigations

Guidance for Industry and FDA Staff Procedures for Handling Post-Approval Studies Imposed by PMA Order

DRAFT GUIDANCE. This guidance document is being distributed for comment purposes only. Document issued on: July 14, 2011

CDRH Device Approval

Distinguishing Medical Device Recalls from Product Enhancements and Associated Reporting Requirements

FDA from a Former FDAer: Secrets and insights into regulatory review and drug development

Draft Guidance for Industry and Food and Drug Administration Staff

Regulatory Issues and Strategies: U.S. FDA

February 15, Re: Docket No. FDA-2017-D-6159: Expedited Programs for Regenerative Medicine Therapies for Serious Conditions

Guidance for Sponsors, Institutional Review Boards, Clinical Investigators and FDA Staff

Biomarker Utility and Acceptance in Drug Development and Clinical Trials: an FDA Regulatory Perspective

Medical Device Accessories: Defining Accessories and Classification Pathway for New Accessory Types. Draft Guidance for Industry and

Guidance for Industry and Food and Drug Administration Staff; The Content of Investigational

Draft Guidance for Industry, Clinical Laboratories, and FDA Staff. In Vitro Diagnostic Multivariate Index Assays

Overview of Biomarker Qualification. Marc K Walton MD, PhD Office of Translational Sciences CDER-FDA

De Novo Classification Process (Evaluation of Automatic Class III Designation) Guidance for Industry and Food and Drug Administration Staff

Document issued on: March 19, The draft of this document was issued on May 20, 2010.

Guidance for IRBs, Clinical Investigators and Sponsors

USE OF PHANTOMS AND IMAGE DATASETS FOR

Draft Guidance for Industry and FDA Staff Premarket Notification [510(k)] Submissions for Medical Devices that Include Antimicrobial Agents

Office for Human Subject Protection. University of Rochester

Docket #: FDA-2018-D-3268

October 13, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

Medical Devices; Exemption From Premarket Notification; Class II Devices; Autosomal

PATHWAYS TO BIOMARKER QUALIFICATION AND ACCEPTANCE

October 25, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

Guidance for Industry ANDAs: Stability Testing of Drug Substances and Products

March 19, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane Room 1061 Rockville, MD 20852

General Comments. May 30, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

GUIDANCE FOR SPONSORS, INDUSTRY, RESEARCHERS, INVESTIGATORS, AND FOOD AND DRUG ADMINISTRATION STAFF

Guidance for Industry and FDA Staff. Replacement Reagent and Instrument Family Policy

Bioresearch Monitoring Inspections in Vitro Diagnostics Devices

Re: Comments on January 2017 Draft Guidance: Considerations in Demonstrating Interchangeability with a Reference Product (Docket No.

Infectious Disease Next Generation Sequencing Based Diagnostic Devices: Microbial

Environmental Assessment: Questions and Answers Regarding Drugs With Estrogenic, Androgenic, or Thyroid Activity Guidance for Industry

FDA identifies this generic type of device as: Absorbable perirectal spacer.

Device Improvements for Pediatric X-Ray Imaging; Public Meeting; Request for Comments

Use of Public Human Genetic Variant Databases to Support Clinical Validity for Genetic

Guidance for FDA Reviewers

The federal food drug and cosmetic Act was originally passed in 1938 and required drugs be proved safe. (click)

Impact of clinical trial design on potential IVD regulatory claims

U.S. FDA CENTER FOR DEVICES AND RADIOLOGICAL HEALTH UPDATE. Jeff Shuren Director Center for Devices and Radiological Health

New and Revised Draft Q&As on Biosimilar Development and the BPCI Act (Revision 2)

Breakthrough Devices Program Draft Guidance for Industry and Food and Drug Administration Staff

Re: Docket No. FDA-2015-D-1246: Draft Guidance on Investigational Enzyme Replacement Therapy Products: Nonclinical Assessment

The Children s Hospital of Philadelphia Committees for the Protection of Human Subjects Policies and Procedures Determination of IND/IDE Requirement

How to put together an IND application

Contains Nonbinding Recommendations. Draft Not for Implementation

DRAFT GUIDANCE. This guidance document is being distributed for comment purposes only. Document issued on: December 27, 2011

Draft Guidance for Industry and Food and Drug Administration Staff Class II Special Controls Guidance Document: External Pacemaker Pulse Generator

Re: Docket No. FDA-2018-D-1895: Indications and Usage Section of Labeling for Human Prescription Drug and Biological Products Content and Format

Compounding Animal Drugs From Bulk Drug Substances; Draft Guidance for Industry;

UT SOUTHWESTERN MEDICAL CENTER AT DALLAS INSTITUTIONAL REVIEW BOARD. Emergency Use of an Investigational Drug, Biologic or Device

Re: Docket No. FDA 2015-D-1580:

DEC DEPARTMENT OF HEALTH & HUMAN SERVICES

DEPARTMENT OF HEALTH & HUMAN SERVICES

ANDREW BALO SENIOR VICE PRESIDENT, REGULATORY, BIOMETRICS, & CLINICAL AFFAIRS DEXCOM, INC 6340 SEQUENCE DRIVE SAN DIEGO, CA 92121

Guidance for Industry and FDA Staff. Criteria for Significant Risk Investigations of Magnetic Resonance Diagnostic Devices

AtriCure Inc. Jonathan McElwee Regulatory Affairs Manger 7555 Innovation Way Mason, Ohio 45040

Deciding When to Submit a 510(k) for a Change to an Existing Device Guidance for Industry and Food and Drug Administration Staff

Guidance for Industry

Compassionate Use Navigator Information for Physicians

BIOMARKERS AND DRUG DEVELOPMENT: REGULATORY PERSPECTIVE

Expanded Access. to Investigational Drugs & Biologics. for Treatment Use

MANUAL: Administrative Policy & Procedure Manual POLICY:

A Risk-based Approach for In Vitro Companion Diagnostics Device FDA Approval Process Associated with Therapies that have Breakthrough Designation

Guidance for Industry DRAFT GUIDANCE

Re: Docket No. FDA-2017-D ; Draft Guidance on Recommendations for Dual 510(k) and CLIA Waiver by Application Studies

BIOMARKERS AND DRUG DEVELOPMENT: REGULATORY PERSPECTIVE

FDA Regulation of Companion Diagnostics

January 3, AtriCure Inc. Melissa Smallwood Associate Regulatory Affairs Specialist 7555 Innovation Way Mason, Ohio 45040

Re: Docket No. FDA 2005-D-0339: Draft Guidance on Drug Safety Information FDA's Communication to the Public

Current Trends at FDA: Implications for Data Requirements

Bipartisan Policy Center, Top Medical Innovation Priorities

Drug Development in Inflammatory Bowel Disease: The FDA Perspective

Disclosures. Laboratory Stakeholders. IVD vs. LDT. FDA Regulation of Laboratory Developed Tests 10/2/2015. FDA Regulation of LDTs

Established Conditions: Reportable CMC Changes for Approved Drug and Biologic Products Guidance for Industry

Guidance for Industry

Guidance. Media Fills for Validation of Aseptic Preparations for Positron Emission Tomography (PET) Drugs DRAFT GUIDANCE

Statement on Regulatory Systems to Improve Pharmaceutical Safety

Use of Real-World Evidence to Support Regulatory Decision-Making for Medical Devices

Structure and Mandate of FDA

Subpart H [Reserved] Subpart I Expanded Access to Investigational Drugs for Treatment. 21 CFR Ch. I ( Edition)

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

December 4, Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852

Docket No. FDA-2011-D-0215 Draft Guidance for Industry and FDA Staff on In Vitro Companion Diagnostic Devices

Contains Nonbinding Recommendations Draft - Not for Implementation

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

Acceptability of Draft Labeling to Support ANDA Approval Guidance for Industry

Evidentiary Considerations for Integration of Biomarkers in Drug Development : Statistical Considerations

May 2, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

RE: Docket No. FDA 2017 N 0041: Agency Information Collection Activities; Proposed Collection; Comment Request; Safety Assurance Case

CDRH s Inspection Strategy for 2018: How it Will Impact Your Company

Unique Device Identification: Convenience Kits Draft Guidance for Industry and Food and Drug Administration Staff

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

Rare Diseases Drug Development and Patient Perspective Initiatives at FDA

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

Guidance for Industry Reference Product Exclusivity for Biological Products Filed Under Section 351(a) of the PHS Act

Guidance for Industry

Guidance for Industry

Transcription:

Reprinted from FDA s website by Medical Device Development Tools Draft Guidance for Industry, Tool Developers, and Food and Drug Administration Staff DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Document issued on: November 14, 2013 You should submit comments and suggestions regarding this draft document within 90 days of publication in the Federal Register of the notice announcing the availability of the draft guidance. Submit written comments to the Division of Dockets Management (HFA-305), Food and Drug Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD 20852. Submit electronic comments to http://www.regulations.gov. Identify all comments with the docket number listed in the notice of availability that publishes in the Federal Register. For questions regarding this document, contact Katie O Callaghan at 301-796-6349 or by electronic mail at kathryn.ocallaghan@fda.hhs.gov. U. S. Department of Health and Human Services Food and Drug Administration Center for Devices and Radiological Health EAS Consulting Group, LLC 1700 Diagonal Road, Suite 750, Alexandria, Virginia 22314 (877) 327-9808 Toll Free (571) 447-5500 Local (703) 548-3270 Fax

Additional Copies Preface Additional copies are available from the Internet. You may also send an e-mail request to dsmica@fda.hhs.gov to receive an electronic copy of the guidance or send a fax request to 301-847-8149 to receive a hard copy. Please use the document number (1882) to identify the guidance you are requesting.

Table of Contents I. INTRODUCTION...1 II. BACKGROUND...2 III. DEFINITION OF KEY CONCEPTS...3 IV. OVERVIEW of CDRH QUALIFICATION POLICY...4 V. CONCEPTUAL FRAMEWORK...5 A. Context of Use...6 B. Tool Types...7 C. Regulatory Considerations and Related Recommendations...11 VI. CONSIDERATIONS FOR QUALIFICATION...11 A. Considerations for CDRH Qualification...12 B. Contents of a Complete Qualification Package...12 VII. CDRH QUALIFICATION PROCESS...15 Stage 1: Pre-Qualification (Optional)...16 Stage 2: Qualification Determination...17 VIII. PROCEDURES FOR SUBMITTING MDDT CORRESPONDENCE AND DOCUMENTS...17 IX. PROCEDURES FOR MAKING QUALIFICATION DECISIONS AVAILABLE...18 APPENDIX 1...20 SAMPLE OUTLINE OF PACKAGE CONTENTS...20

Draft Not for Implementation 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 Medical Device Development Tools Draft Guidance for Industry, Tool Developers, and Food and Drug Administration Staff This draft guidance, when finalized, will represent the Food and Drug Administration's (FDA's) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance. I. INTRODUCTION This document provides draft guidance on a voluntary process for qualification of medical device development tools (MDDT) for use in device development and evaluation programs in the Center for Devices and Radiological Health (CDRH). CDRH believes that application of this policy will facilitate the development and timely evaluation of innovative medical devices, by providing a more efficient and predictable means for collecting the necessary information to make regulatory assessments. The purpose of this guidance is to describe the framework and process for voluntary CDRH qualification of MDDT, including definitions of applicable terms, criteria for evaluating an MDDT for a specific context of use, considerations for qualification, and the contents of a qualification submission. This guidance does not discuss the review of MDDTs submitted as part of a premarket regulatory submission for a specific medical device, nor does it address the specific evidentiary or performance expectations FDA would have for the qualification of a specific MDDT. This draft guidance applies to both therapeutic and diagnostic devices unless otherwise specified. FDA's guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency's current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.

41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 II. BACKGROUND An MDDT is a scientifically validated tool a clinical outcome assessment (e.g. patient-reported or clinician-reported rating scales), a test used to detect or measure a biomarker (e.g. assay for a chemical analyte or medical imaging method), or non-clinical assessment method or model (e.g. in vitro, animal or computational model) - that aids device development and regulatory evaluation. Qualification reflects CDRH s expectation that within a specified context of use 1, the results of an assessment that uses an MDDT can be relied upon to support device development and regulatory decision-making. The intent of this voluntary CDRH qualification policy is to (1) enable faster, more efficient development of important life-saving and health promoting medical devices, (2) promote the development of tools to facilitate more timely device evaluation, (3) provide a mechanism to better leverage advances in regulatory science, and (4) more quickly and more clearly communicate to stakeholders about important advances in regulatory science that may be leveraged to speed device development and regulatory evaluation. We expect the qualification process to expedite development of publicly available tools which could potentially be used widely in multiple device development programs. Once an MDDT is qualified for a specific context of use, FDA s expectation is that it can be used by any medical device developer for that context of use. CDRH reviewers should accept the MDDT for the qualified context of use without the need to reconfirm the suitability of the MDDT. Importantly, the existence of a qualified MDDT does not convey a requirement that the tool must be used during the device development or regulatory evaluation process. Other scientifically valid tools or approaches may also be used. CDRH is committed to advancing regulatory science, which provides the tools, standards, and approaches needed to evaluate the safety, effectiveness, quality, and performance of the products we regulate. Through continued advances, such as this qualification process, we are modernizing the regulatory evaluation process and reducing the time and resources needed to develop and assess new products. This promotes innovation, supports the manufacture of high quality products, and speeds the rate at which safe and effective technologies reach the market. 1 For the purposes of this guidance, the term context of use refers to a key aspect of qualification -- the use parameters for which the MDDT has been validated. This use is defined in part by the device or product area in which the MDDT can be qualified, the stage of device development, and the specific role of the MDDT (for clinical uses, this includes the study population or disease characteristics). The context of use defines the boundaries within which the MDDT is qualified. 2

74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 III. DEFINITION OF KEY CONCEPTS For the purposes of this guidance, the following definitions apply: A Medical Device Development Tool (MDDT) is a scientifically validated tool - a clinical outcome assessment (e.g. patient-reported or clinician-reported rating scales), a biomarker test (e.g., assay for a chemical analyte or medical imaging method), or nonclinical assessment method or model (e.g. in vitro, animal or computational model) that aids device development and regulatory evaluation. Qualification is a conclusion that within a specified context of use, CDRH expects that the results of an assessment that uses an MDDT can be relied upon to support device development and regulatory decision-making. Context of use refers to a key aspect of qualification. This use is defined in part by the device or product area for which the MDDT is qualified, the stage of device development, and the specific role of the MDDT (for clinical uses, this includes the study population or disease characteristics, as well as specific use diagnosis, patient selection, clinical endpoints). The context of use defines the boundaries within which the MDDT is qualified. A Clinical Outcome Assessment (COA) relies on subjective measures of how a patient feels or functions, and is sometimes used to determine whether or not a device demonstrates a treatment benefit. COAs include patient-reported, clinician-reported, and observer-reported outcome measures and are typically instruments composed of a scale or score. A patient-reported outcome (PRO) assessment captures the patient perspective concerning symptoms or functioning. A clinician-reported outcome assessment is based on clinical observation or interpretation by a trained clinician. An observer-reported outcome is assessed by observers without the need for clinical expertise. Examples of COA include: pain scales, quality of life or health status scores, NIH Stroke Scale. Other clinical outcomes based on subjective clinical decision-making may also be qualified as MDDTs if they may be used as a measure of treatment benefit when clearly defined. Examples include: heart failure-related hospitalization or reoperation rate. A Biomarker Test (BT) is a test or instrument (e.g. an in vitro/laboratory test or medical imaging method) or other objective measurement method used to detect or measure a biomarker. A biomarker is a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or responses to a therapeutic intervention. 2 Examples of BT include: an instrument or method for 2 Modified from: Biomarkers Definitions Working Group (2001). Clinical Pharmacology and Therapeutics, 69, p. 89 95. 3

113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 measuring blood pressure (e.g., sphygmomanometry); an instrument or method for measuring certain concentrations of serum proteins. A Nonclinical Assessment Model (NAM) is a nonclinical test method or model used in device development or evaluation that reflects device function or in vivo performance. A NAM could be an in vitro ( bench ) model, animal model, or computational model and is developed to measure a parameter of interest or to substitute for another generally accepted test or measurement. Examples of NAM include: in vitro models to replace animal testing, the use of tissue and other material phantoms to evaluate imaging devices, electromagnetic phantoms, and validated computational models. IV. OVERVIEW OF CDRH QUALIFICATION POLICY Qualification reflects CDRH s expectation that within the specified context of use, the results of an assessment that uses an MDDT can be relied upon to support medical device development and regulatory decision-making. Once qualified, CDRH expects that the MDDT may be used by device developers for the qualified context of use in regulatory submissions without the need for CDRH review staff to reconsider and reconfirm the suitability of the MDDT with each submission. Medical device developers may use qualified MDDTs, but are not required to do so. Why is CDRH developing a qualification process? This qualification process provides a mechanism for leveraging advances in regulatory science, fostering MDDT development and adoption, and facilitating faster, more efficient device development and regulatory evaluation. This voluntary process can facilitate the scientific evaluation and assessment of a medical device by providing a more efficient and predictable means for collecting the necessary information to make regulatory assessments. Qualification, as described in this guidance, is intended to increase efficiency in the device development process by providing some degree of generalizability for use of MDDTs across multiple medical types or clinical disorders, to advance device development and more widely benefit patients. The extent of generalization will depend on the MDDT, as well as the strength of evidence and justification for a broad proposed context of use. Qualification also facilitates collaboration in a pre-competitive setting where multiple interested parties (individuals, companies, or organizations) may work together to develop an MDDT for qualification. This may result in a reduction in the resources expended by each individual collaborator and motivate interested parties to join an MDDT development effort, thus expediting the MDDT development and use. CDRH encourages the formation of collaborative 4

153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 groups to foster MDDT development programs to increase the efficiency of tool development through joint efforts and to lessen the resource expenditure of any individual person or company. As further detailed in Section VII, we are available to discuss potential MDDT development programs with stakeholders. A qualification process may have advantages for CDRH as well. Previously, if there was interest in using a particular MDDT for multiple products or different clinical settings, each FDA device review team would typically evaluate the data justifying the MDDT use for each product or setting separately. Instead, if an MDDT is qualified through the process described in this guidance, the relevant data supporting the tool would be reviewed thoroughly during this process, so that the MDDT could be relied upon within the qualified context of use in the future, without redundant, detailed review of the suitability of the test. What does CDRH Qualification mean for regulatory decision-making? The decision to qualify an MDDT means that after reviewing relevant available scientific evidence, CDRH intends to consider the MDDT a valid tool within the defined context of use and to rely on assessments using the MDDT for regulatory purposes. Qualification decisions will be made public and reflect CDRH s support for the general use of the MDDT within the specified context of use, not just for a single, specific device submission. The value to the public health will be increased as new MDDTs become widely known and available for use by multiple device developers. A qualification decision involves a consideration of (1) the specified context of use; (2) the strength of available evidence supporting the MDDT (including tool validity, plausibility, etc); and (3) an assessment of the advantages and disadvantages of relying on assessments using the MDDT within the specified context of use. MDDT qualification does not obviate the need for a device developer to meet existing regulatory requirements or alter the benefit-risk threshold for regulatory decision-making related to a medical device; rather, it can facilitate the scientific evaluation and assessment of a medical device by providing a more efficient and predictable means for collecting the necessary information to make regulatory assessments. The CDRH premarket review divisions maintain responsibility for evaluating new devices using information obtained using a qualified MDDT. V. CONCEPTUAL FRAMEWORK CDRH has defined three types of MDDTs: COAs (including patient- and clinician-reported outcomes), BTs (such as assay or medical imaging methods), and NAMs (such as in vitro, animal or computational models). Each type of MDDT may have a variety of potential uses relevant to CDRH s regulatory evaluation of new medical devices. MDDTs are instruments, tools or methods of measurement that address outcomes and are subject to quality issues such as 5

196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 accuracy, precision, reliability, reproducibility. These quality issues may affect the usefulness of an MDDT. The following sections provide more detail about context of use, distinguish this from MDDT type, and outline certain regulatory considerations for distributing and using qualified MDDTs. A. Context of Use The context of use refers to a key aspect of qualification. It describes the way the MDDT should be used and the purpose of the use. Once an MDDT is qualified, the context of use defines the boundaries within which the available data adequately support use of the MDDT. Context of use is defined in part by 1) the device or product area for which the MDDT is qualified, 2) the stage(s) of device development (e.g., early feasibility study, pivotal study, etc.), and 3) the specific role of the MDDT (for clinical uses this includes the study population or disease characteristics, as well as specific use diagnosis, patient selection, clinical endpoints). Different categories of contexts of use for an MDDT: 1. Aid in Diagnosis As a definition of an adverse event (AE) within a clinical study As a clinical reference standard to assist in diagnosis 2. Patient Selection 3 For selection of clinical trial subjects To stratify patient population by predicted risk 3. Clinical Endpoints 4 As an intermediate endpoint 5 3 MDDTs need not be FDA cleared or approved products. However, when an MDDT test is used in a clinical trial as a companion diagnostic, in that it is essential for the safe and effective use of a corresponding developed therapeutic product, it must comply with applicable investigational use requirements. Developers may wish to see FDA s draft guidance In Vitro Companion Diagnostic Devices for FDA s proposed approach on this topic. (available at http://www.fda.gov/medicaldevices/deviceregulationandguidance/guidancedocuments/ucm2 62292.htm). 4 A clinical endpoint is a detected symptom or measurement of a function, or any clinical characteristic or variable that reflects how a patient feels, functions, or survives, measured at a specific timepoint. Examples: mortality reports; loss of sight. 6

223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 As a surrogate endpoint 6 4. Non-clinical Device Assessment Bench or animal study methodologies which reduce test duration or minimize sample size As a substitute for an evaluation typically conducted through human or animal studies Reliance on in vitro or in silico studies to reduce or minimize the use of animals The MDDT may also have potential value outside these boundaries. The MDDT may be used in device development programs for a different purpose other than the qualified context of use, subject to review and discussion with CDRH on a case-by-case basis. In addition, the qualified context of use for the MDDT may be expanded over time as additional data are obtained. If data become available that call into question the validity, appropriateness, or assessment of advantages and disadvantages of a previously qualified context of use, CDRH may modify or withdraw the qualification. B. Tool Types CDRH recognizes three types of MDDT, distinguished primarily by how the tool measures relevant parameters. Tools that measure clinical parameters via some subjective metric are Clinical Outcome Assessments (COA). Tools that measure clinical parameters via an objective approach (e.g., physical measurement or chemical analysis) are considered Biomarker Tests 5 An intermediate endpoint is itself a clinical endpoint concerning a symptom or measure of function that is not the ultimate outcome of the disease. Improvement according to an intermediate endpoint is of value to patients even if this does not lead to reduced morbidity or mortality. An intermediate endpoint may also be a clinical endpoint measured at an earlier timepoint than has historically been accepted. A treatment effect shown by an intermediate endpoint may also be taken as reason to expect a favorable ultimate outcome; in this sense the intermediate endpoint plays the role of a surrogate. For example, exercise tolerance is sometimes used as an intermediate endpoint in trials of treatments for heart failure. 6 A surrogate endpoint is a measurement used in trials as a substitute for a clinical endpoint, and is expected to reflect clinical outcomes based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence. For example, blood pressure measurements are sometimes used as endpoints in trials of antihypertensive therapeutics, as a surrogate for clinical endpoints of stroke, myocardial infarction, or mortality. 7

248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 (BT). Tools that measure nonclinical parameters are categorized as Nonclinical Assessment Models (NAM). These MDDT types are further detailed below. 1. Clinical Outcome Assessment A Clinical outcome assessments (COAs) is a subjective measures of how a patient feels or functions, and is sometimes used to determine whether or not a device demonstrates a treatment benefit. COAs include patient-reported, clinician-reported, and observerreported outcome measures and are typically instruments composed of a scale or score. A widely used example is the NIH Stroke Scale. The reporter (i.e. clinician, patient, or other observer) of the outcome distinguishes the type of COA. A clinician-reported outcome (ClinRO) assessment is based on clinical observation and interpretation by a trained clinician. An observer-reported outcome (ObsRO) is assessed by observers without the need for clinical expertise. Patient reported outcomes (PROs) 7 are a common subtype of COA. They are a measurement of the patient s health condition based on a direct patient (i.e., study subject) report without amendment or interpretation by someone else. A COA includes not only the measure that produces a score but also the clearly defined methods and instructions for administration of the tool, a standard format for data collection, and well-documented methods for scoring, analysis, and interpretation of results in the targeted patient population. COAs can measure treatment benefit directly (e.g., a PRO for pain intensity) or indirectly (e.g., a diary of rescue pain medication use for pain intensity). Qualification of a COA as an MDDT includes a review of the evidence that the proposed tool is a valid assessment for how patients feel or function in day-to-day activities. Other clinical outcomes based on subjective clinical decision-making may also be qualified as MDDTs if they may be used to assess a treatment benefit. Examples may include hospitalization rate or reoperation rate. CDRH intends to qualify a COA based on a determination that for a specified context of use, assessment of a clinical outcome using the COA provides valid scientific evidence when used in a well-controlled investigation (see 21 CFR 860.7). 7 Issues relevant to FDA review of both new and existing PROs are summarized in FDA s guidance for industry on Patient-Reported Outcome Measures: Use in Medical Product Development to Support Labeling Claims (http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ UCM193282.pdf). Many of the issues described in that guidance are also relevant to ClinROs and ObsROs. 8

283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 2. Biomarker Test A biomarker is a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or responses to a therapeutic intervention. A biomarker can be a physiologic, pathologic, or anatomic characteristic or measurement that relates to an aspect of normal or abnormal biologic function or process. Typically, a biomarker is measured using a test (e.g., by chemical analysis) or instrument (e.g., by sphygmomanometry). CDRH intends to consider MDDTs derived from medical imaging to be BTs, if the characteristic (e.g., tumor diameter) is objectively measured. As with other MDDTs, CDRH intends for qualified BTs to be relied upon to support device-related regulatory decision-making for the defined context of use. BTs may be used to select patients for inclusion in a device clinical trial, to monitor treatment response, to predict or identify safety problems related to treatment with a medical device, or to identify patients who are or are not candidates for certain forms of therapy. Appropriate use of qualified BTs may increase the efficiency of the device development and evaluation process by providing reliable predictions about device performance. Fundamentally, in order for a BT to be useful, it must be sufficiently accurate and precise. When considering BTs for qualification, the evaluation will assess both the strength of evidence supporting the biomarker for the specified context of use, as well as the validity of the test instrument and/or methodology to measure the biomarker. In qualifying a BT, FDA implicitly accepts the strength of evidence supporting the biomarker for the specified context of use. Subsequent BTs seeking qualification for similar contexts of use need only demonstrate the validity of the test instrument and/or methodology to measure that same biomarker. The amount and type of new evidence needed to support qualification of a BT will depend on whether there is any test or instrument for measurement of the biomarker that is already FDA cleared or approved for clinical use through premarket review, and if so, whether the proposed context of use for the BT is consistent with the indication for use of the cleared or approved product. To qualify a BT to measure a biomarker for which there is no corresponding FDA cleared or approved device, test validity (e.g., precision and accuracy) should be demonstrated in the proposed context of use. In contrast, MDDT qualification proposals involving an FDA cleared or approved test, or involving biomarkers for which FDA has established the analytical performance criteria necessary for measurement for the specified context of use, can make use of existing data that support the analytical validity of the test, so long as the MDDT submitter is legally authorized to do so. In either case, qualification depends on meeting performance criteria for the test or instrument s accuracy, precision, etc. as an MDDT in the proposed context of use. 9

326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 Importantly, the processes and criteria for qualification of a BT as an MDDT remain distinct from requirements for pre-market product review, even when the products (biomarker test for use in device development and legally marketed medical device), the objects of the product (biomarker and analyte) and inferences drawn (biology and clinical significance) are similar or identical. However, information developed for pre-market review, such as analytical validity, may be potentially relevant to qualification of a BT, even when the proposed context of use differs from the cleared or approved indication for use of the device. The MDDT submission should clearly identify the elements of BT qualification (Section VI) that are addressed using information from a premarket submission. MDDT qualification determinations will in no way affect the regulatory or compliance status of any product intended for commercial distribution (see Section C). 3. Nonclinical Assessment Model A nonclinical assessment model (NAM) is a nonclinical test method or model that reflects device function or in vivo performance and is used in device evaluation to measure a parameter of interest, or to substitute for another generally accepted test or measurement. NAM examples include in vitro models that replace or minimize the need for animal testing; the use of tissue and other material phantoms to evaluate imaging devices; validated computational models; and the development and validation of a new animal model to evaluate a device in lieu of clinical data. Qualified NAMs may be used to evaluate a new material property, modifications to an existing design, or a device feature historically evaluated through animal or human testing. The MDDT qualification process and standards recognition process 8 may be viewed as complementary. We anticipate that NAM qualification may be most useful for models or methods which are not yet covered by standards or guidance. In some cases, an MDDT which is addressed in an FDA-recognized consensus standard may have already been assessed in a manner similar to the MDDT qualification process and qualification for the same context would likely not be beneficial. However, for those that have not, assessments conducted during development of the standard might contribute evidence toward MDDT qualification (e.g., round-robin testing conducted by the standards organization to assess tool validity). In addition, models proposed for different contexts of use or which necessitate different or more specific methodology than described in existing standards could also potentially be qualified through the MDDT process. 8 Information on CDRH s standards program is available at http://www.fda.gov/medicaldevices/deviceregulationandguidance/standards/default.htm. 10

366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 C. Regulatory Considerations and Related Recommendations Some MDDTs may meet the definition of a device in section 201(h) of the FD&C Act and be subject to the statutory and regulatory requirements applicable to devices, such as 510(k) clearance or premarket approval. Devices intended for investigational use are exempt from most of these requirements, including clearance and approval, as long as there is compliance with applicable investigational use requirements, such as those under 21 CFR part 812. A qualified MDDT typically would be used in the research or investigation of a medical device, according to the MDDT s context of use. Such use would generally exempt a qualified MDDT that is a device from clearance, approval, and other device requirements, as long as a clinical investigation meets applicable requirements: If the device that is the subject of the investigation is a significant risk 9 device as defined by 812.3(m), the investigation, including use of the MDDT, is subject to all requirements of 21 CFR 812. If the device that is the subject of the investigation is not a significant risk device as defined by 812.3(m), the abbreviated requirements listed under 812.2(b) apply to the investigation, including the use of the MDDT. Investigations, including the use of the MDDT, that meet the criteria for one of the exemptions described in 812.2(c), including 812.2(c)(3), are not required to comply with part 812 with the exception of 812.119. For qualified MDDTs that are devices that are not cleared, approved, or 510(k)-exempt, any statement that the device has been qualified by FDA should be accompanied by a disclaimer that qualification of an MDDT does not constitute FDA clearance or approval. Without such a disclaimer, the labeling could be considered misleading, which would render the device misbranded under sections 201(n) and 502(a) of the FD&C Act. If an MDDT appears to be a device under the FD&C Act, we will discuss these issues with you during the qualification process. VI. CONSIDERATIONS FOR QUALIFICATION As stated previously, CDRH s qualification decision means that CDRH expects that within the specified context of use, an MDDT can be applied in device development (to evaluate potential device designs in clinical or nonclinical settings) and that the results of an assessment that uses 9 A determination about risk of the investigation should be made (see CDRH guidance: http://www.fda.gov/downloads/regulatoryinformation/guidances/ucm126418.pdf). 11

404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 an MDDT can be relied upon to support regulatory decision-making (to support marketing applications and labeling modifications). A. Considerations for CDRH Qualification When determining whether to qualify a proposed MDDT, CDRH intends to consider the key aspects listed below. The amount and strength of evidence needed to support qualification depends largely on the defined context of use. Description of MDDT. Is the MDDT adequately described? Context of use. Is the context of use adequately and appropriately defined? Strength of evidence. Does the available scientific evidence demonstrate that the MDDT reliably and accurately measures what it is intended to measure, is scientifically plausible, and is reasonably likely to predict the outcome of interest? Assessment of advantages and disadvantages. Within the specified context of use and given the available strength of evidence, do the advantages of using the MDDT outweigh potential disadvantages of making decisions based on measurements obtained using the MDDT? B. Contents of a Complete Qualification Package 1. Description of MDDT The qualification package should specify the MDDT type (COA, BT, or NAM), describe the measurements provided by the MDDT, and provide a descriptive summary of the MDDT principle and methodology of measurement. 2. Context of Use The qualification package should describe how and where the MDDT would be used within the device evaluation program. This includes 1) the device or product area in which the MDDT is proposed t be qualified, 2) the stage of device development (design evaluation, animal testing, early clinical study, pivotal clinical study to support market application, non-clinical PMA data requirement, post-market design or label changes), and 3) specific role of the MDDT (for clinical uses, this includes the study population or disease characteristics, as well as specific use diagnosis, patient selection, clinical endpoints). For examples, please refer to section V.A. 12

446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 3. Strength of Evidence The qualification package should discuss the strength of evidence for the MDDT, and address the following areas: Tool Validity. Does the available data adequately support the validity of the measurement? Does the MDDT measure reliably and accurately? Depending on the tool type, this may include analytical, clinical, and construct validity, sensitivity, specificity, accuracy, precision, repeatability, external validity, reduction of bias, verification of the constitutive model, uncertainty quantification, numerical convergence, etc. Plausibility. Is it scientifically plausible that the measurements obtained through use of the MDDT are related to the true outcome of interest? Is there a causal path or mechanistic explanation to connect the MDDT to the outcome? Extent of Prediction. What data are available to demonstrate a predictive relationship between the MDDT and the true outcome of interest? What is the strength of that predictive relationship? Is the prediction repeatedly demonstrated in multiple studies or as a class effect? If relevant, is the conclusion (that the effect of treatment on the measurement obtained using the MDDT predicts the outcome of interest) supported by credible information? 10 Capture. Does the MDDT fully capture the aggregate effect of the intervention on the true outcome of interest? Does the MDDT account for every major effect of the intervention? Are there available data which call this into question? The amount and strength of evidence needed to support qualification of an MDDT will vary depending on the context of use and the MDDT type. For example, an MDDT proposed for use as one component of a definition of an adverse event (AE) within a clinical study may need less data compared to a BT proposed for use measuring a primary endpoint for a pivotal study to evaluate a novel high-risk device type; the latter use may need more evidence of its validity due to the potential disadvantages of accepting an inaccurate MDDT in this context. 10 For the purposes of this guidance, credible information includes data generated under the design control procedures of 820.30, nonclinical or animal testing, peer reviewed published literature, or other reliable information such as clinical information gathered during a trial or marketing. This definition is consistent with FDA s use of the term in Part 812. 13

483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 4. Assessment of Advantages and Disadvantages As part of the qualification determination, CDRH intends to consider an assessment of advantages and disadvantages for qualification of the MDDT. The qualification package should discuss the advantages and disadvantages of accepting the MDDT. CDRH intends to consider the following factors: Assessments of Advantages of Using the MDDT: This should take into account the following factors: o The type of advantage(s). Advantages may include: significantly accelerating the time to develop and evaluate devices; allowing for shorter or smaller clinical or nonclinical studies; allowing for safer or less invasive, easier, more convenient, or less variable measurements than the alternative; and expediting the development of a novel technology of public health importance. o Magnitude of advantage. This may include: whether there is a potential to impact multiple device development programs; whether the context of use includes life-threatening and/or serious chronic diseases or conditions, or diseases/conditions where there are no or poor alternatives; or whether the MDDT is to be used for novel technology where there is no established paradigm for regulatory assessment. o Likelihood of an advantage. This is based on the strength of evidence (tool validity, plausibility, correlation/prediction, capture) in support of the MDDT, and a comparison to the available alternatives. Assessments of Disadvantages of Using the MDDT: This should take into account the following factors: o Type(s) of risks. Considering the context of use, what types of decisions might be made based on the use of an MDDT that should not have been qualified? These are considered within the context of use, including: 1) the device or product area in which the MDDT is proposed to be qualified, 2) the stage of device development (design evaluation, animal testing, early clinical study, pivotal clinical studies to support market application, postmarket design changes); and 3) the specific role of the MDDT (for clinical uses, this includes the study population or disease characteristics, as well as specific use diagnosis, patient selection, clinical endpoints). 14

526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 o Magnitude of risk. The scope of impact of making a decision based on inaccurate conclusions from an MDDT is based on the severity of risk, a comparison of the MDDT to its alternatives, and considering the context of use. o Likelihood of risk. How likely is a particular risk to occur? This could be based on the evidence in support of tool validity. For a diagnostic test, this could be the likelihood of the MDDT reporting a false positive, false negative or false estimate of predictive value. o Risk mitigation. The use of mitigations may minimize the risks of relying on the MDDT. For example, alternative sources of information or confirmatory data from later timepoints may mitigate risks of decision-making based on information from an MDDT. Additional Factors for Assessing Advantages and Disadvantages of Using the MDDT: The following factors may apply: o Degree of certainty. If the advantages of using the MDDT are high, less certainty (less rigorous strength of evidence) may be acceptable to support its use. On the other hand, if the advantages are minimal, or if the potential disadvantages are great, more rigorous evidence may be needed to support MDDT use. o Novelty of technology. The assessment will consider whether MDDTs facilitate development and regulatory evaluation of devices that address areas of unmet need, or that incorporate new technologies (especially first-of-a-kind) which may offer advantages that did not previously exist. Particularly where providers and patients have limited alternatives available, MDDT use may facilitate patient access and encourage innovation. 5. Consent to Public Disclosure and Use In order to obtain FDA qualification, MDDT submitters must provide authorized consent (1) for FDA to make public sufficient information to support use of the qualified MDDT and (2) for the general public to use the MDDT and rely on data generated using the MDDT in gaining FDA clearance or approval of other devices. VII. CDRH QUALIFICATION PROCESS During the CDRH process for MDDT qualification the Agency and MDDT submitters interact to efficiently determine the amount and type of information needed to support qualification for a 15

568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 specific tool and context of use. The qualification process consists of two stages: 1) an optional pre-qualification stage, and 2) a qualification determination stage. Throughout the pre-qualification and qualification determination stages of review, CDRH intends to prioritize proposals for evaluation of the MDDT according to the following factors: Public health need met by one or more of the following: o Context of use includes life-threatening and/or serious chronic diseases or conditions; o No/poor alternatives or unmet scientific need; o Novel or innovative technology with no established paradigm for regulatory assessment; o Major efficiencies to be gained in device development and evaluation time. Scope of impact: o Potential to impact multiple device development programs; o Potential to impact multiple sponsors. The number of proposals accepted for detailed CDRH involvement will depend on available resources. Where appropriate, CDRH may seek input from external individuals or groups for specific expertise, consistent with all applicable statutory and regulatory requirements, including those respecting confidentiality. Once an MDDT is qualified for a specific use, the context of use may be modified or expanded over time in response to new data or changing science. Modification or incremental expansion of the qualified context of use over time may be undertaken through the qualification process. Alternatively, if the growing body of scientific evidence no longer supports the context of use, CDRH may withdraw the MDDT qualification. Stage 1: Pre-Qualification (Optional) The process for MDDT qualification can be triggered in one of 3 ways: 1) FDA identifies an area of need and/or calls for development activity in a specific area; 2) need and interest in an area is determined by individual or consortia of stakeholders (may include academia, industry, medical societies); 3) a MDDT developer chooses to pursue qualification for its tool to allow for broad use across multiple device development programs. Prioritization of Proposals Interested parties should submit a proposal including a concise overview of the qualification project, and description of the need for the MDDT (see Appendix 1). Priority proposals may be accepted for early direct FDA staff involvement. Consultation and Evidence Development 16

611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 The qualification review team (which may include FDA as well as external expertise, where appropriate) should interact with the submitter to identify the amount and type of data or information needed for qualification of the proposed MDDT for the context of use. CDRH intends to notify applicants of whether they have been selected for a pre-qualification meeting or teleconference with FDA staff. Additional interactions or correspondence should occur as needed during the MDDT development stage. Stage 2: Qualification Determination When the submitter has the data and information necessary for a complete qualification package, they may submit it to justify qualification of the MDDT for the proposed context of use (see Appendix 1). The qualification review team should interact with the submitter as needed for clarification or to request additional information. CDRH intends to hold a qualification meeting or teleconference to facilitate discussion once the package has been reviewed. In the case of complex or controversial MDDT programs, CDRH may seek external expertise or public comment. In evaluating an MDDT for qualification, CDRH would not consider whether there may be restrictions on use of the tool stemming from patent. CDRH does not have the resources or the expertise to review patents and individual patent claims, or otherwise be involved in issues related to patent law. Upon completion of the evaluation, CDRH intends to notify the submitter in writing of the qualification determination. VIII. PROCEDURES FOR SUBMITTING MDDT CORRESPONDENCE AND DOCUMENTS All MDDT correspondence and documents for CDRH should be clearly labeled as a MDDT qualification submission, and sent to the Document Control Center (DCC). 11 Submitters should include an ecopy 12 as well. 11 Submissions to CDRH should be sent to: U.S. Food and Drug Administration, Center for Devices and Radiological Health, Document Mail Center WO66-G609, 10903 New Hampshire Avenue, Silver Spring, MD 20993-0002. 12 For more information on formatting of an ecopy, please see:http://www.fda.gov/downloads/medicaldevices/deviceregulationandguidance/guidancedo cuments/ucm313794.pdf. Although submission of an ecopy for an MDDT Qualification Submission is voluntary, if you choose to submit an ecopy, it should meet the technical standards outlined in Attachment 1 of the referenced guidance. 17

643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 The Cover Letter should contain the following elements: Date: Subject: (in bold print) MDDT QUALIFICATION SUBMISSION MDDT Type: (in bold print) o CLINICAL OUTCOME ASSESSMENT, o BIOMARKER TEST, or o NONCLINICAL ASSESSMENT MODEL MDDT Tracking Record Number: (in bold print), if previously assigned Submission Type: (in bold print) o REQUEST FOR PRELIMINARY DISCUSSION or PRE- QUALIFICATION PROPOSAL or o INTERIM CORRESPONDENCE or o QUALIFICATION PACKAGE. MDDT Name(s): (in bold print): Identify the specific MDDT (by name) that is being submitted Context of Use: Describe the intended context of use of the MDDT (1 to 2 sentences) Complete submitter contact information including name(s), affiliation, mailing address, email address, phone and fax numbers IX. PROCEDURES FOR MAKING QUALIFICATION DECISIONS AVAILABLE 18

675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 To make information about qualified MDDTs available to the public, CDRH intends to use the following process: To allow for public comment for each new qualification determination, FDA intends to publish a draft appendix to this guidance and issue a notice of availability of new and/or revised (draft) qualification determinations. FDA expects the notice to identify a comment period for draft determinations. Once finalized, FDA intends to publish the qualification determination as an appendix to this guidance. CDRH expects to announce MDDT qualification determinations on an established MDDT Web page. With permission from a submitter, CDRH also intends to post new MDDTs in the process of developing evidence to support qualification, so that any parties interested in participating can contact the submitter. CDRH intends to provide detailed supporting documentation and information, when appropriate, on the MDDT Qualification Web page, or in some cases (e.g., for certain NAM) to capture that information in a master file for reference by multiple sponsors. CDRH expects to make public sufficient information to support broad use of the qualified MDDT. In order for submitters to particulate in this voluntary qualification process, they must agree that information about the qualified MDDT will be made publicly available for use in device development programs in the specified context of use. To this effect, as appropriate, CDRH intends to provide information for the public about how to access the MDDT. 19

699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 APPENDIX 1 SAMPLE OUTLINE OF PACKAGE CONTENTS The recommended outline below is provided as an example only for illustrative purposes and is not required. A sample package may include the following information: (1) a description of MDDT type, principle and methodology; (2) context of use; (3) amount and strength of available evidence; (4) assessment of advantages and disadvantages for qualifying the MDDT; and (5) consent to public disclosure and use. 1. Description of MDDT MDDT Type: o Clinical Outcome Assessment (COA) o Biomarker Test (BT) o Nonclinical Assessment Model (NAM) Description of measurements provided by the MDDT: o What does the MDDT measure or provide? o Is it intended to replace a previously accepted measurement? Descriptive summary of the MDDT principle and methodology of measurement 2. Context of Use Device or product area in which the MDDT can be qualified Stage of device development o Design evaluation o Animal testing o Early clinical study o Pivotal clinical study to support marketing application o PMA nonclinical data requirement o Post-market design or label changes Specific role of the MDDT: Non-clinical Device Assessment o Bench or animal studies methodologies which reduce test duration or minimize sample size o As a substitute for an evaluation typically conducted through human or animal studies 20