临床试验总结报告英文版

临床试验总结报告英文版
临床试验总结报告英文版

Reviewer: Cynthia A. Rask, M.D.

Through: Marc K. Walton, M.D., Ph.D.

Karen Weiss, M.D.

Date: 23 January 2002

Biologics License Application Supplement

Medical Officer’s Review

Rebif? (Interferon β-1a)

FOR TREATMENT OF

RELAPSING-REMITTING MULTIPLE SCLEROSIS

Submitted by Serono, Inc.

STN #: 103780 / 0

TABLE OF CONTENTS OVERVIEW (4)

Scope of this revie w (4)

Abbreviations and Definitions of Terms Used in This Review (5)

INTRODUCTION (7)

Multiple Sclerosis (7)

Background and Diagnostic Criteria (7)

Current Treatment of MS (8)

The Orphan Drug Act and Orphan Drug Exclusivity Regulations (8)

Overview of Prior Clinical Studies of Rebif? (10)

PROTOCOL XXXXXXXXXX (12)

Objectives (12)

Design (12)

Material Source (13)

Randomization (14)

Inclusion Criteria (14)

Exclusion Criteria (14)

Treatment (15)

Evaluations Performed During the Study (17)

Efficacy Endpoints (21)

Primary Endpoint (21)

Secondary Endpoints (21)

Tertiary Endpoints (21)

Safety Endpoints (21)

Safety Measurements Analyzed (21)

Statistical Analysis Plan (22)

Determination of Sample Size (22)

Analysis Populations (22)

Planned Interim Analysis (23)

Significance Testing, Allocation of Alpha (23)

Analysis of Baseline Parameters (23)

Primary Endpoint – Proportion of Subjects Exacerbation-Free at 24 Weeks (24)

Secondary Endpoints – MRI Analytic Methods (24)

Safety Analyses (26)

Study Administration (26)

STUDY RESULTS (27)

Formal Protocol Modifications (27)

Changes in the Conduct of the Study or Planned Analyses (27)

Protocol Deviations/Violations (28)

Study Conduct at Specific Study Sites (30)

Subject Enrollment and Disposition (31)

Randomization (31)

Time on Study (31)

Time on Treatment: (32)

Adverse Events Leading to Premature Discontinuation (33)

Demographics and Baseline Characteristics (35)

EFFICACY RESULTS (38)

Primary Endpoint Results (38)

CBER-Performed Analyses on the Primary Endpoint (42)

Secondary Endpoints (48)

MRI-Determined CU Activity (48)

Exacerbation Rate per Subject (49)

Mean Number of T2 Active Lesions per Subject per Scan (50)

Tertiary Endpoints (51)

Exploratory Analyses (53)

Safety Analyses (55)

Serious Adverse Events and Deaths (55)

Important Rare Serious Adverse Events (58)

Severe Adverse Events (59)

Analysis by Body System and Event (60)

Pregnancies (62)

Development of Antibodies to Interferon-? (62)

Assessment and Conclusions (63)

Financial Disclosure Statements (65)

References (65)

Overview

Interferon ? -1a (Rebif?) has been under development by Serono, Inc. for the treatment of multiple sclerosis (MS). Serono, Inc. submitted a Biologics License Application (BLA) on February 27, 1998 for the approval of Rebif? for the treatment of patients with relapsing-remitting MS (RRMS). The focus of the application was a 560-subject study that was a randomized, double-blind, placebo-controlled study of 22 μg vs. 44 μg Rebif? vs. placebo administered subcutaneously (SC) three times per week for 2 years. The application also contained additional open-label safety data from other studies. Based on the review of the BLA submission conducted by CBER, it was concluded that both doses of Rebif? were demonstrated to be safe and effective and to be approvable for treatment of RRMS. However, Serono was prevented from marketing Rebif? in the United States by the Orphan Drug Exclusivity previously granted to Biogen for the marketing of their interferon β-1a, Avonex? and to Berlex/Chiron for the marketing of their interferon β-1b, Betaseron? Betaseron?’s exclusivity expired in July 2000. Rebif? was, however, successfully approved and marketed in other countries, and thus has been commercially available in other portions of the world for several years. The subject of the current BLA amendment is a comparative study that Serono conducted to compare the efficacy of Rebif? to marketed doses of Avonex? in the treatment of relapsing-remitting MS. The study design was discussed with CBER and the design was agreed upon prior to initiation of the study. The study was a randomized, open-label study in which subjects with RRMS were treated with either Rebif? 44 μg SC three times per week or Avonex? 30 μg IM once weekly, with neurologic examinations performed by physicians blinded to the treatment assignment and with subjects’ MRIs read at a central reading facility by neuroradiologists blinded to treatment assignment. Although the duration of the study was to be 48 weeks, the pre-specified primary outcome measure was the proportion of subjects who remained relapse-free following 24 weeks of treatment. Supportive evidence (secondary endpoints) were comparisons of MRI abnormalities. The Applicant conducted and submitted this BLA amendment in the belief that demonstration of superior clinical benefit of Rebif? over Avonex? would allow them to break Biogen’s orphan drug exclusivity and thus, to market Rebif? in the U.S. for the treatment of relapsing-remitting MS.

Scope of this review

The focus of this document is upon efficacy information obtained primarily from a single study, XXXXXXXXXX, a randomized, open-label comparative study of the use of Rebif? 44 μg administered SC 3x per week vs. Avonex? 30 μg administered IM once weekly. Efficacy and safety data from the BLA previously submitted by the Applicant and reviewed by CBER for the treatment of relapsing-remitting MS will be summarized. Safety data from several additional studies of Rebif? in MS, including safety data from the Applicant’s post-marketing safety database will also be summarized.

Abbreviations and Definitions of Terms Used in This Review

2-5 OAS 2’, 5’-oligoadenylate synthetase

ADL Activities of Daily Living

AE Adverse Event

ALT Alanine Transaminase

ANOVA Analysis of Variance

ANCOVA Analysis of Covariance

AST Aspartate Transaminase

Avonex? Biogen’s recombinant human interferon β-1a BBB Blood-brain barrier

Betaseron? Berlex’s human interferon β-1b

CHO Chinese Hamster Ovary

CRA Clinical Research Associate

CRF Case Report Form

CU Combined Unique (T1 + T2)

DER Drug Even t Report (form)

EC Ethics Committee

ECG Electrocardiogram

EDSS (Kurtkze’s) Expanded Disability Status Scale Gd Gadolinium

Gd-DPTA Gadolinium-diethylenetriaminepentacetic acid HCG Human Chorionic Gonadotrophin

HLA Human Lymphocyte Antigen

HTLV-1 Human T-cell lymphotrophic virus, type 1 IEC Independent Ethics Committee

IFN Interferon

IFN β-1a Recombinant human interferon β-1a

IM Intramuscular (ly)

IRB Institutional Review Board

IU International Unit(s)

IUD Intrauterine device

IV Intravenous (ly)

KFS Kurtzke Functional Systems

mcg microgram

mg milligram(s)

mL milliliter

MIU Million International Units (106 IU)

MR Magnetic Resonance

MRI Magnetic Resonance Imaging

MS Multiple Sclerosis

MSCRG Multiple Sclerosis Collaborative Research

Group

NSAID Non-steroidal anti-inflammatory drug

PRISMS Prevention of Relapses and disability by

Interferon β-1a Subcutaneously in Multiple

Sclerosis

prn as needed

qod every alternate day

qw once weekly

Rebif? Serono’s recombinant human interferon β-1a

RRMS Relapsing-remitting multiple sclerosis

SAE Serious Adverse Event

SC Subcutaneous (ly)

SGOT (AST) Serum glutamic oxaloacetic transaminase

SGPT (ALT) Serum glutamic pyruvic transaminase

SNRS Scripps Neurological Rating Scale

SPECTRIMS Secondary Progressive Efficacy Clinical

Trial of Recombinant Interferon-beta in

Multiple Sclerosis

SPMS Secondary progressive multiple sclerosis

T1 T1-weighted MRI scanning sequence

T2 T2-weighted MRI scanning sequence

tiw three times per week t 1/2half-life

μg microgram(s)

ULN Upper Limit of Normal

USAN United States Adopted Names

USP United States Pharmacopoeia

WHO World Health Organization

WHOART World Health Organization Adverse

Reaction Terminology

Introduction

Multiple Sclerosis

Background and Diagnostic Criteria

Multiple Sclerosis (MS) is a chronic, inflammatory, demyelinating disease of the central nervous system of unknown etiology, although an autoimmune process has often been implicated. It is a common cause of neurological disability in young adults, primarily affecting people between 20 and 40 years of age, and affects women approximately twice as often as men. Although there are sometimes inconsistencies of terminology with regard to categorization of MS within the MS field, and although some persons with MS do not neatly fit into one of the following categories, it has often proved useful to discuss MS according to clinical course, in order to: 1) provide advice on prognosis, 2) incorporate this information into consideration of potential therapeutic choices, and 3) to improve homogeneity and thus chances for detecting clinically meaningful effects in designs of clinical trials of new therapies. Most experts in the field generally recognize three clinical forms of MS: relapsing-remitting, secondary progressive and primary progressive (Lublin and Reingold, 1996). Relapsing-remitting MS (RRMS) comprises the most common form at onset. It is the presenting form in up to an estimated 80-85% of subjects, and involves recurrent attacks of neurological symptoms and signs (relapses or exacerbations) involving multiple areas of the nervous system that occur at variable time intervals ranging from months to years between attacks. These exacerbations or relapses are followed by subsequent variable degrees of recovery (remissions). The majority of subjects with RRMS develop secondary progressive MS (SPMS) in which periods of stable recovery give way to neurological decline over time. About 50% of subjects with RRMS will develop the secondary progressive form of the disease within 10 years of onset; the proportion approaches 80% after 25 years (Runmarker and Anderson, 1993). Primary progressive MS is distinct from SPMS and is characterized by steady accumulation of neurological deficit from onset, without superimposed attacks or exacerbations; it affects a much smaller percentage of subjects, perhaps 10-20% of subjects with MS present with this form.

Subjects with secondary progressive MS may continue to experience relapses; such subjects are then sometimes referred to as suffering relapsing-progressive disease; however, others (Weinshenker et al., 1989), use the term relapsing progressive MS to define subjects with a category they regard as exhibiting a mixture of primary progressive and secondary progressive disease, not simply a subset of secondary progressive MS. Such disparities in the use of terminology such as relapsing progressive account for some of the confusion in the literature regarding categorizations of MS beyond the three widely accepted categories of relapsing-remitting, secondary progressive and primary progressive disease. All experts agree, however, that there is a general tendency for the frequency of relapses to decline with time as the slowly progressive nature of the disease becomes more prominent. Weinshenker et al. in 1989 were amongst the first to provide comprehensive natural history data that supported the belief that progression of disability following conversion to SPMS is more rapid than prior to conversion.

Several phases of pathological change occur in MS, including breakdown of the blood-brain barrier with edema, lymphocytic infiltration with cytokine release, demyelination and axonal loss. Functional impairment can occur with any of these pathological processes, but irreversible deficits are thought to result from demyelination and axonal injury. There is evidence from magnetic resonance spectroscopy (Arnold et al., 1994 and DeStefano et al., 1998) and pathological studies (Trapp et al., 1998 and Ferguson et al., 1997) that axonal dysfunction and loss can occur even at early clinical stages of the disease.

Current Treatment of MS

There are currently four drugs approved for the treatment of MS in the United States. Betseron? (Interferon β-1b) and Avonex? (Interferon β-1a) have been demonstrated to have beneficial effects on relapsing-remitting MS. Betaseron? administered at a dose of 8 MIU SC every other day was demonstrated to decrease the frequency of clinical exacerbations in ambulatory patients with relapsing-remitting MS. Avonex? administered at a dose of 30 μg IM once weekly was demonstrated to decrease the frequency of clinical exacerbations and to slow the accumulation of physical disability as measured by Kurtzke’s Expanded Disability Status Scale (EDSS) score. Copaxone? (glatiramer acetate – formerly known as copolymer-1) administered at a dose of 20 μg SC once daily was demonstrated to decrease the frequency of relapses in patients with relapsing-remitting MS. All three drugs have consistently been shown to reduce the frequency of clinical exacerbations compared to placebo by approximately one-third. Their effects have never previously been compared directly in a clinical trial. Novantrone? (Mitoxantrone), a cancer chemotherapeutic agent, was approved in 2000 for reducing neurologic disability and/or the frequency of clinical relapses in patients with secondary (chronic) progressive, progressive relapsing, or worsening relapsing-remitting multiple sclerosis. However, its cumulative dose-limiting cardiotoxicity restricts its role in the treatment of MS.

Other immunosuppressive agents, including cyclophosphamide, azathioprine and low-dose methotrexate have been demonstrated to have only modest effects in treating MS, and are not widely used in this country. Similarly, intravenous immunoglobulin infusions (IVIG) are felt by some to be effective in treating MS, but are not widely used in the U.S., and do not have an approved indication for the treatment of MS.

The Orphan Drug Act and Orphan Drug Exclusivity Regulations

The Orphan Drug Regulations in 21 CFR Part 316 were written to implement section 2 of the Orphan Drug Act of 1983, that consists of four sections added to the Federal Food, Drug, and Cosmetic Act. The Orphan Drug Act directs FDA to provide written recommendations on studies required for approval of a marketing application for an orphan drug. It also provides for the designation of drugs, including antibiotics and biological products, as orphan drugs when certain conditions are met, and it provides conditions under which a sponsor of an approved orphan drug enjoys exclusive approval for that drug for the orphan indication for 7 years following the date of the drug’s marketing approval.

The orphan drug regulations state that “after approval of a sponsor’s marketing application…FDA will not approve another sponsor’s marketing application for the same drug before the expiration of 7 years from the date of such approval” (21CFR316.31(a)). The definition of “exclusive approval” clarifies that this exclusivity applies only to the same indication as the originator product’s approval (21CFR316.3(b)(12)).

The definition of “same drug” (21CFR316.3(b)(13)) describes how to judge two products from different sponsors on a physical-chemical basis for different classes of molecules. For each description, however, a showing that the follow-on product is clinically superior to the originator product can supercede the structural determination of “same drug”.

The definition of “clinically superior” (21CFR316.3(b)(3)) describes the criteria for judging the subsequent product clinically superior to the already approved product. A new product can be deemed clinically superior if it has shown greater effectiveness on a clinically meaningful endpoint. For this demonstration, “in most cases, direct comparative clinical trials would be necessary.” Alternatively, showing greater safety is adequate to deem a product clinically superior, with only “in some cases, direct comparative clinical trials” needed for this demonstration. Additionally, “in unusual cases, a demonstration that the drug otherwise makes a major contribution to patient care” can be adequate to deem the second product clinically superior to the already approved product.

In 1993 Berlex Laboratories, in conjunction with Chiron, obtained marketing approval for their interferon β-1b product, Betaseron?, for treatment of relapsing-remitting MS. Berlex had previously obtained orphan drug designation for Betaseron? for treatment of MS, so that a 7-year period of marketing exclusivity for Betaseron? began with its approval. The clinical efficacy claim of the indication was for a decrease in the frequency of clinical exacerbations in ambulatory patients with relapsing-remitting MS.

In 1996 Biogen, Inc. obtained marketing approval for their interferon β-1a product, Avonex? for treatment of relapsing forms of MS. Biogen had previously obtained orphan drug designation for Avonex? for treatment of MS. During the review of Biogen’s PLA for Avonex?, CBER determined that under the orphan drug regulations Betaseron? and Avonex? were deemed the same drug for purposes of determining marketing exclusivity when physical structure alone was considered. However, the two products were deemed different when the clinical data were considered. Avonex? treatment was found to have a significantly different and superior safety profile with regard to the incidence of injection-site skin necrosis, and was thus concluded to be a different drug from Betaseron? for orphan drug marketing exclusivity purposes. Consequently, Avonex? received marketing approval in 1996 and its own 7-year period of marketing exclusivity from its approval date. The clinical efficacy claim of Avonex?’s indication was to slow the accumulation of physical disability and decrease the frequency of clinical exacerbations in patients with relapsing forms of MS.

Like Berlex and Biogen, Serono, Inc. was granted orphan drug designation for Rebif? for the treatment of MS. When they submitted a BLA in January of 1998 for the approval of their interferon β-1a product, Rebif?, for the treatment of relapsing-remitting and “transitional”

MS, CBER determined that by structural comparison only, Rebif? was deemed to be the same product as either Betaseron? or Avonex? for orphan drug purposes, since two protein products that differ only in minor amino acid sequences or in glycosylation of the product are explicitly stated in the orphan drug regulations to be regarded as the same product. Consequently, clinical data remained the only mechanism by which Rebif? could possibly be determined to be not the “same drug” for orphan drug marketing exclusivity purposes. Following their review of the data submitted by Serono in the BLA filed in 1998, CBER concluded that Rebif? should not be given marketing approval while the existing marketing exclusivity accorded to Betaseron? (expired in 2000) and Avonex? (expires in 2003) remains in effect based on the following conclusions:

FDA determined that on a solely structural basis, Rebif?, Betaseron? and Avonex? would be deemed the same drug, making clinically based information the only avenue for distinguishing th e drugs for use within the currently approved patient population. Consequently, there was no basis for determining that Rebif? is not the same drug as either Avonex? or Betaseron?, since

? Serono had not submitted any information addressing a clinical comparison of Rebif? to Betaseron?;

? Serono had not submitted adequate evidence to demonstrate that Rebif? has superior clinical efficacy or superior safety over Avonex?;

? Serono had not submitted adequate evidence to demonstrate that Rebif? makes a major contribution to patient care over Avonex?;

? Serono had not submitted adequate evidence to demonstrate that a medically plausible subset of subjects with MS, distinct from those for whom Avonex? is indicated, has been studied and shown to benefit from Rebif? where no benefit may be expected from

Avonex?.

Although MS is no longer considered an orphan disease today because the prevalence is now known to be more than 250,000 in the U.S., the Orphan Drug Exclusivity regulations regarding marketing exclusivity still apply to the beta-interferons, since they were granted orphan drug status for the treatment of MS at a time when MS was estimated to affect fewer than 200,000 persons in the U.S.

Overview of Prior Clinical Studies of Rebif?

Serono has completed 6 controlled trials in MS along with a number of uncontrolled trials. Two studies were previously reviewed here in CBER in 1998-1999 as part of Serono’s initial BLA application for the approval of Rebif? for treatment of relapsing-remitting MS. Study GF 6789 was a 560-subject, multicenter, randomized, placebo-controlled trial that compared two doses of Rebif? (22 μg or 44 μg) vs. placebo administered SC three times per week for 2 years. The primary endpoint was the number of protocol-defined exacerbations per subject using the following definition of an exacerbation: “the appearance of a new symptom or worsening of an old symptom, attributable to MS, accompanied by appropriate new neurological abnormality, or focal neurological dysfunction lasting for at least 24 h ours in the absence of fever, and preceded by stability or improvement for at least 30 days.” Rebif?

treatment significantly reduced the number at one and two years of treatment and the efficacy did not differ substantially between the two doses, nor was treatment with Rebif? associated with excessive numbers of dose-limiting toxicities. Rebif? treatment also showed a positive treatment effect on many of the secondary endpoints studied (not listed in order of importance by the Applicant prospectively), including effects on moderate/severe exacerbations, time to first exacerbation, proportion of subjects exacerbation-free, time to confirmed disability and percent progressors, decrease in MRI T2 lesion volume, decrease in combined T1 and T2 MRI “activity,” and reduction in hospitalizations and steroid treatment for MS. However, it was noted in the review that the incidence of “troubling” adverse events that included cytopenias and hepatic enzyme abnormalities were notably higher for the 44 μg dose than the 22 μg dose and both were higher than the rates observed in the placebo group. The percentages of these adverse events are shown in Table 1.

Table 1: Cytopenias and Liver Enzyme Elevations in Study GF 6789

WHOART preferred term Placebo

N=187 IFN 22

μg TIW

N=189

IFN 44

μg TIW

N=184

lymphopenia 11.2% 20.1% 28.8% leukopenia 3.7% 12.7% 22.3% granulocytopenia 3.7% 11.6% 15.2% thrombocytopenia 1.6% 1.6% 8.2%

SGPT increased 4.3% 19.6% 27.2%

SGOT increased 3.7% 10.1% 17.4%

hepatic function abnormal 1.6% 3.7% 9.2%

It was also noted in the review that one subject had developed a severe anaphylactoid reaction that was deemed probably due to the administration of Rebif?. It occurred approximately 1 month after treatment was begun and progressed over several days to “localized symptoms of urticaria, generalized pruritus and swollen red scars at the injection site.” Subsequent skin tests were said to have shown that the subject was allergic to a component in both the active drug and placebo. There was no increase in the incidence of depression or suicidal ideation or attempt in either treatment group compared to placebo. It was concluded that hematologic and hepatic toxicity was noticeably increased in a dose-dependent manner. Some of these abnormalities resulted in decisions to stop treatment. Although severe hematopoietic events occurred with greater frequency in active treated groups, there was no increase in infections. Hepatic enzyme abnormalities appeared to be isolated laboratory abnormalities. It w as also noted that there were noticeable increases of important injection site adverse events with Rebif? therapy, but rare discontinuation of treatment due to these events.

A post-marketing report of “severe anaphylaxis with recombinant interferon β1a,” Rebif?, was submitted as a letter to the Editor in Neurology in 1999.

XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX XXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXXX. Protocol XXXXXXXXXX

Title: An Open-Label, Randomized, Multicenter, Comparative, Parallel Group Study of Rebif? 44 μg Administered Three Times per Week by Subcutaneous Injection, Compared with Avonex? 30 μg Administered Once per Week by Intramuscular Injection in the Treatment of Relapsing-Remitting Multiple Sclerosis

Period of Study Conduct: November 1999 to February 2001

Funding: Serono, Inc.

Objectives

The primary objective as stated by the Applicant in the protocol was to demonstrate that the proportion of patients with relapsing-remitting MS who were exacerbation-free would be greater with Rebif? 44 μg administered three times per week (132 μg per week) than patients treated with Avonex? 30 μg once per week for 24 weeks.

The principal secondary objective as stated in the protocol was that the MRI-determined combined unique (CU) lesion activity would be less after 24 weeks of treatment with Rebif? 44 μg three times per week than with Avonex? 30 μg once per week.

Design

This was a multicenter, open-label, randomized, comparative, parallel group study in which up to 624 interferon-na?ve subjects with relapsing-remitting multiple sclerosis (RRMS) were to be equally randomized to receive either Rebif? 44 μg administered SC three times per week or Avonex? 30 μg administered IM once per week for 48 weeks. Although all enrolled subjects were to complete 48 weeks of the treatment to which they were randomized, the efficacy outcomes were to be determined after 24 weeks of treatment.

T2-weighted and T1-weighted pre- and post gadolinium enhanced MRIs were obtained within 28 ± 4 days of beginning treatment and monthly thereafter following the start of treatment until Week 24. All pre- and post-treatment MRIs were read centrally at the

XXXXXXXXXX, where the raters received only the subject’s identification number, initials, date of birth and whether or not the subject was to be scanned again.

Each center was required to have two separate physicians responsible for the management of each subject: a treating physician and an evaluating physician. The treating physician was responsible for the supervision of study drug administration, for reporting and treating adverse events and monitoring safety assessments, and was also responsible for the treatment of exacerbations and for determining whether non-MS-related factors could account for neurological worsening. The evaluating physician was responsible for neurological assessments and evaluation of exacerbations, and was responsible for determining whether an apparent exacerbation met the protocol’s definition. The evaluating physician was to remain unaware of treatment assignments, adverse event profiles and any changes in safety assessments throughout the trial. Both the treating physicians and subjects were instructed not to discuss these issues with the evaluating physician.

The primary study endpoint was the proportion of subjects who were exacerbation-free at 24 weeks. Subjects were instructed to inform the study center within 48 hours of the onset of an exacerbation, and at that time, the treating physician or designate would discuss the symptoms with the subject and determine whether a neurological examination was indicated. If so, the subject would be advised to come to the center for evaluation. The evaluating physician was to determine whether or not an exacerbation meeting the protocol’s definition had occurred, and would evaluate its severity based on changes in the EDSS and the Kurtzke Functional Systems (KFS) score.

If the treating physician determined that corticosteroids were necessary for treatment of the exacerbation, a standard regimen of 1.0 g/day of IV methylprednisolone for three days was to be administered.

Treatment discontinuation was mandatory in case of any of the following:

1. Pregnancy

2. Loss to follow-up

3. Use of other investigational products, or

4. Use of other approved disease-modifying therapy for MS

Treatment could be discontinued for other reasons, but subjects were asked to continue the study assessments if they discontinued from the study prematurely.

Material Source

Rebif? was supplied as a sterile solution in pre-filled syringes for subcutaneous administration. Each syringe contained 0.5 mL of solution, which consisted of 44 μg (12 MIU) of interferon β-1a, 4 mg albumin (human, USP), 27.3 mg mannitol (USP), water for injection (USP) and XXXXXXXXXX for pH adjustment. The batch numbers used in this study were: XXXXXXXXXX.

Commercially available Avonex? 30 μg for IM administration was reconstituted and administered according to the directions in the package insert. The batches used were: XXXXXXXXXX.

Randomization

Treatment assignments were determined using a computer-generated randomization list generated by the Serono Biometrics Department and were allocated through a centralized telephone randomization system. Randomization was stratified by center, with an initial block size of six followed by block sizes of four. This was done, according to the Applicant, after they performed simulations that determined the randomization block size that best prevented the potential detection of the treatment assignment for the next subject to be enrolled within a center. Randomization was to have occurred within 24 hours of a subject completing screening procedures and having been found eligible for the study.

Inclusion Criteria

Subjects were deemed eligible for participation in the study if they met the following criteria:? Age between 18 and 55 years

? Clinically definite or laboratory-supported diagnosis of relapsing-remitting MS, according to Poser’s criteria

? Two or more relapses within the preceding 24 months

? Clinical stability or improving neurological state during the four weeks prior to Study Day 1

? EDSS score of 0 to 5.5, inclusive

? Two or more lesions consistent with MS on a screening T2-weighted MRI performed within 28 ± 4 days of Study Day 1

? Adequate contraception for females of childbearing potential

Exclusion Criteria

Subjects were to be excluded if any of the following were present:

? Secondary progressive, primary progressive or progressive relapsing MS

? Prior use of interferon

? Treatment with oral or systemic corticosteroids or ACTH within 4 weeks of Study Day 1 or 7 days of the screening MRI

? Significant leukopenia, defined as a white blood cell count of <0.5 x the lower limit of normal within 7 days of Study Day 1

? Elevated liver function tests (ALT, AST, alkaline phosphatase or total bilirubin > 2 x the upper limit of normal) within 7 days of Study Day 1

? Prior cytokine or anti-cytokine therapy or glatiramer acetate within the 3 months prior to Study Day 1

? Immunomodulatory or immunosuppressive therapy within the 12 months before Study Day 1, including, but not limited to, cyclophosphamide, cyclosporin, methotrexate,

azathioprine, linomide and Mitoxantrone

? Previous use of cladribine or total lymphoid irradiation

? Allergy to human serum albumin, mannitol or Gd-DTPA

? IV immunoglobulin or any other investigational drug or procedure in the 6 months before Study Day 1

? Systemic disease that could interfere with subject safety or evaluation of their MS Treatment

Dose and Administration

Subjects enrolled in this study were to receive one of two treatments for a period of at least 48 weeks:

? Rebif? 44 μg, administered SC three times weekly, or

? Avonex? 30 μg, administered IM once weekly

Rebif? was supplied as a sterile solution in pre-filled syringes. Subjects were advised to dose in the evening to minimize the impact of any adverse event and were instructed to dose each Monday, Wednesday and Friday. Instructions for self-administration, including instruction in proper injection techniques, advice regarding rotation of injection sites and advice about avoiding injection into inflamed areas was provided prior to the start of Rebif? treatment.

Avonex? was supplied in its commercially available form: a lyophilized powder to be reconstituted with sterile water for injection. Subjects were advised to administer Avonex? by following the instructions in the package insert. Subjects were not advised as to day of the week or time of day to administer Avonex? in the protocol, but individual investigators were able to advise subjects of a recommended dosing schedule.

In Amendment #4 to the protocol dated November 9, 2000, subjects were given the option of using an auto injector to self-administer their Rebif? after completion of the 24 Week Visit. Dose Titration

In order to minimize potential side effects at the beginning of treatment with Rebif?, a dose titration schedule was instituted. The dose administered was gradually increased over the first four weeks of treatment, with 8.8 μg 3 times per week (20% of total) for the first two weeks, 22 μg 3 times per week (50% of total) for the third and fourth weeks, and the full dose of 44 μg 3 times per week was given thereafter.

The Avonex? dose was not titrated, and was administered at 30 μg once per week beginning at Study Day 1 and continued throughout the study to subjects randomized to that treatment arm.

Dose Modification for Toxicity

Toxicity was to be graded by the treating physician according to the Modified WHO Recommendations for the Grading of Acute and Subacute Toxicities. Dose modification was not to occur in case of neurological events.

In case of Grade 1 or 2 toxicity, subjects were to be treated as deemed appropriate by the treating physician. If Grade 2 toxicity persisted, the dose of Rebif? could be reduced to 50%, but the full dose was to be resumed if at all possible.

In case of Grade 3 toxicity attributable to Rebif?, the dose could be reduced to 50% or 20% as the treating physician deemed appropriate, or interrupted until the toxicity resolved to Grade 0 or 1. An attempt to resume the full dose was to be made within 4 weeks by escalating to the next dose level (50% or 100%). If Grade 3 toxicity recurred, the dose could again be reduced or interrupted until resolution to Grade 0 or 1 and then resumed or maintained at 50% dose for the remainder of the study. Further recurrence of Grade 3 toxicity or persistence after 4 weeks’ interruption would result in permanent discontinuation of treatment.

In case of Grade 4 toxicity attributable to Rebif?, subjects were to be withdrawn from treatment.

For subjects randomized to the Avonex? treatment arm, investigators were told to follow the instructions supplied in the package insert for Avonex? to make adjustments for toxicity. Investigators could also consider the use of recommendations for Rebif? dose modifications if needed.

Concomitant Therapy

Treatment of Relapses During the Study

If the treating physician determined that corticosteroids were necessary for treatment of a clinical exacerbation of the subject’s MS, a standard regimen of 1.0 g/day of IV methylprednisolone for three days was to be administered. If the subject were due to have a study-related MRI scan, the scan would be performed either before beginning methylprednisolone or at least 7 days after the last dose.

Treatment of Flu-Like Symptoms

Acetaminophen (paracetamol) could be given prophylactically or to treat constitutional symptoms associated with the study drugs, such as fever, myalgia or influenza-like symptoms. Non-steroidal anti-inflammatory drugs could be given if acetaminophen did not relieve these symptoms or if a subject could not tolerate acetaminophen.

Treatment of Other Symptoms

Except for those listed in the study Exclusion Criteria, any medication that was considered necessary for a subject’s welfare and that would not interfere with study treatment could be given at the treating physician’s discretion and recorded in the Case Report Forms. Treatment Compliance

Subjects were provided with diary cards on which they were to record the v olume of each dose, the time of administration, adverse events, and any concomitant medications. These cards were collected and reviewed by the investigator at each study visit. The subjects also used drug accountability records to document the dispensing of study medications and the return of unused drugs and empty containers.

Evaluations Performed During the Study

The study flowchart, based on the September 30, 1999 version of the protocol is shown in Table 2.

Non-laboratory screening procedures were to have been performed within 28 ± days before the planned start of treatment, after informed consent had been obtained. Laboratory screening procedures were to have been performed within 7 days before the planned start of treatment.

On Study Day 1, the first day of study treatment, a complete physical and neurological examination, including evaluation of the subject’s EDSS score, KFS score, ambulation distance and timed ambulation were to have been performed within 24 hours of the planned treatment initiation. The evaluating physician was not to refer to the subject’s examination results before performing the examination. Proton density T2-weighted, pre-Gd T1-weighted and post-Gd T1-weighted MRI scans were also to be obtained at this visit; these scans were to have been separated from the screening MRIs by at least 24 hours, but not more than 32 days, and could be performed up to 3 days prior to the initiation of treatment.

Table 2: Study Procedures

Procedure 28

Days

Prior

to

Day

1 7

Days

Prior

to

Day

1

Study

Day 1

Week

4

Week

8

Week

12

Week

16

Week

20

Week

24

Every

12

Weeks

Medical

History,

Physical

Examination*

X X X X X

Minor Office

Visit

X X X X

Neurological

Examination**

X X X X X MRI X X X X X X X X X****

Labs*** X X X X X Thyroid

X X X**** Function

Tests

X X X**** Antibodies to

IFN-β

Document

X X X X X X X X X Exacerbations

X X X X X X X

Adverse

Events

Concomitant

X X X X X X X X X

Medications

*after screening, includes only weight and vital signs

** includes the EDSS, KFS, Distance Walked and Timed Ambulation Index

*** includes hematology, blood chemistries, urinalysis

****MRIs, thyroid function tests and antibodies to IFN-β to be done at Weeks 48, 72, etc. in extension study

Neurological Examinations

Measures were undertaken to try to keep the examining physician, who was to perform all neurological examinations, blinded to treatment assignment. Subjects were instructed not to disclose their treatment or anything related to th eir treatment regimen, to cover injection sites before neurological examinations, and not to discuss symptoms that might be related in any way to their study treatment, such as injection site reactions or influenza-like symptoms, with the evaluating physician. Evaluating physicians were instructed to communicate with subjects only about neurological matters, and to remind subjects not to discuss other matters related to treatment. Neurological examinations included evaluations of the EDSS, KFS scores, ambulation up to 500 meters and timed ambulation (evaluation of subjects walking a measured distance of 8 meters or 25 feet along a straight course as fast as they were able – the time taken was recorded). All neurological examinations were to be performed without consulting a subject’s previous neurological examination.

When neurological examinations and MRI scans were scheduled for the same visit, they were to be performed on the same day whenever possible; otherwise a time difference of no more than ± 48 hours between the evaluations was permissible.

Evaluation and Treatment of Exacerbations During the Study

An exacerbation was defined as the appearance of a new symptom or worsening of an old symptom attributable to MS, accompanied by an appropriate new n eurological abnormality or focal neurological dysfunction lasting at least 24 hours in the absence of fever, and preceded by stability or improvement for at least 30 days. An “appropriate new neurological abnormality” was defined as one that was consistent with the new neurological symptoms reported by the subject, whether or not accompanied by new findings in other systems. “Focal neurological dysfunction” was defined as a symptom of CNS disturbance, possibly accompanied by objective neurological findings (such as a change in sensory perception with myelitis or change in visual acuity/color perception with optic neuritis), which was felt to be consistent with an MS exacerbation. Subjects were instructed to inform the study center

within 48 hours of the onset of an exacerbation, and at that time, the treating physician or designate would discuss the symptoms with the subject and determine whether a neurological examination was indicated. If so, the subject would be advised to come to the center for evaluation. The evaluating physician was to determine whether or not an exacerbation meeting the protocol’s definition had occurred, and would evaluate its severity based on changes in the EDSS and the Kurtzke Functional Systems (KFS) score, as follows:

? Mild: EDSS change of 0 to 0.5 point, with a new neurological finding and/or and increase in KFS score of one point in one to three systems

? Moderate: EDSS change of 1.0 to 2.0 points and/or an increase in KFS score of one point in four or more systems or of two points in one to three systems

? Severe: EDSS and/or KFS score increases that exceed those described for a moderate exacerbation

The evaluating physician was not to refer to the subject’s previous examination(s) before performing the neurological examination for determination of an exacerbation, but if necessary, was allowed to review the subject’s previous results in order to determine if the criteria for an exacerbation had been met, and if so, the exacerbation severity. If the EDSS and KFS score changes were disparate, the exacerbation was to be graded according to the greater severity.

The treating physician would determine appropriate management of the exacerbation, including whether to prescribe corticosteroid treatment, and would perform a separate assessment of exacerbation severity based on effects on the activities of daily living (ADL), using the following scale:

? Mild: little or no effect on the ADLs

? Moderate: significant impact on the ADLs

? Severe: need for hospitalization for treatment or management of the exacerbation

The treating physician or designate would conduct weekly phone checks to determine when the exacerbation reached maximum severity and began to improve. If necessary, the subject would undergo further neurological examinations; exacerbation severity would be graded according to the worst EDSS and KFS scores recorded during the exacerbation.

Subject Contacts Between Scheduled Visits

These contacts were to have occurred by telephone at Weeks 2, 6, 10, 14, 18 and 22 to determine whether any symptoms consistent with an exacerbation had occurred and whether or not an unscheduled neurological examination by the evaluating physician was needed. Unscheduled Visits

Subjects could be seen at any time during the study for evaluation of a possi ble MS exacerbation or for evaluation of possible adverse events.

Procedures for and Evaluation of MRI Scans

MRIs were performed in accordance with a scanning protocol that was designed to standardize procedures across all centers. The scanning protocol specified positioning techniques, the expected range of scanning parameters, and the exact content and format required on the film and computer tapes to be sent for central analysis at the MS/MRI

Analysis Group at the XXXXXXXXXX. The MS/MRI Analysis Group had no clinical knowledge of any of the subjects whose scans they analyzed. Scan data were read, converted, and stored in the XXXXXXXXXX MS/MRI Analysis Group’s computer for quantitation and analysis. All films and tapes were checked for completeness, consistency, and compliance with the MRI scanning protocol. Scans that did not meet the standards set in the protocol were rejected for analysis. Throughout the study, communication was maintained between the MRI scanning sites and the MS/MRI Analysis Grou p, with frequent feedback to the sites concerning the quality and consistency of scans.

Assessment of T2 Activity

A T2 active lesion was defined as any new, recurrent, newly enlarging or persistently enlarging T2 lesion; a T2 active scan was defined as a scan showing any T2 active lesions. ? New T2 lesions were those that had not appeared in any previous scan.

? Recurrent T2 lesions were those appearing at a site where earlier lesions had disappeared. ? Enlarging T2 lesions were those showing identifiable increase in size following previous stability; they cold be either newly enlarging, if enlargement was not observed on the previous scan, or persistently enlarging, if enlargement had also been noted on the

previous scan

Assessment of T1 Activity

A T1 active lesion was defined as any newly enhancing, recurrent enhancing or persistently enhancing T1 lesion; a T1 active scan was defined as a scan showing any T1 active lesions. ? Newly enhancing T1 lesions were those that had not enhanced in any previous scan. ? Recurrent enhancing T1 lesions were enhancing lesions appearing at a site where earlier lesions had disappeared.

? Persistently enhancing T1 lesions were those that had enhanced on previous scans and continued to enhance on the current scan.

Assessment of Combined Unique Activity

A combined unique (CU) active lesion was defined as any lesion that was T1 active, T2 active or both; a CU active scan was defined as a scan showing any CU active lesions. Combined unique lesion counts were obtained in three steps:

? T1 active lesions were identified by sequential review of the subject’s T1 scans and each was assigned a contrast identification number that was recorded in a database.

? Similarly, T2 active lesions were identified by sequential reviews of T2 scans, and each was assigned a morphological identification number.

? The two sets of scans (T1 and T2) were then reviewed together. When a T1 active lesion and a T2 active lesion were determined to be the same, the contrast and morphological identification numbers were linked to avoid double counting of simultaneous activities in single lesions. Links could involve the current, previous, or subsequent scan. Numbers of non-linked T1 active lesions, non-linked T2 active lesions and linked lesions showing both T1 and T2 activity were then combined to give counts of new CU active lesions (consisting of new CU and recurring CU lesions) and persistent CU active lesions. Adverse Event Collection

临床试验总结报告的体例格式和内容要求内容

临床试验总结报告体例和容要求 1.题目封面 封面题页应包括如下容: ——试验题目 ——试验药物/研究产品的名称 ——试验用药品的适应症 ——如不能在题目中表明、则简要描述(以1~2句话)设计(平行、交叉、双盲、随机)比较(安慰剂、活性成分、剂量/反应),间隔,剂量和病例数。 ——申办者 ——原始规划与计划的确认(编码或数目,确认日期) ——试验预期进度与进程安排 ——研究开始数据(受试的第一位病人,或任何其它数据) ——后期研究终末数据(最终一个受试者的名称、或终末研究者完成的数据) ——试验研究完整的数据(最后的受试者) ——参加者或合作者或主办者责任医务官等人士的名称和职责。 ——药业公司名称/主办方签名(在公司/主办者中负责研究报告的人,公司/主办方联络者的,和传真,以备在回顾研究报告提出问题时,在此页码或应用的字母有所表明)。 ——表明该项研究是否在优良临床试验管理规(GCP)条件下进行的包括应达到的基本文件、研究设施、人员知识结构,统一培训等。 ——报告的资料(通过题目和资料,确定与该项研究相同的任何其它较早报告) 2.容概述 应提供—个临床试验的主要容提要(通常限于3页容),该提要应包括试验题目、研究人员、研究出版物名称、试验时间、试验目的、试验方法、试验研究样本、诊断及进入研究的主要标准、试验的产品、剂量、给药方式及批号、治疗持续时间、参考的治疗、剂量、给药方式及批号、评价标准(有效性、安全性)、统计方法、总结——结论(效能结论、安全性结论、结论)及报告日期。该提要应包括表明结果的数字资料,而不仅仅是文字和P值(见附件l,研究容概述)。 3.容目录 容表格应包括: ——页码或其它每一个部分的局部资料,包括归纳性表格、图和图表。 ——附录、列表和任何能提供的病例报告形式的汇总和定位。 4.名词、术语缩写和定义 应提供报告中使用的—系列缩写符、特殊的或不常用的术语,或测量单位定义,应拼写出缩写术语,并在文中首次出现时表明其全称。

医疗器械临床试验病例报告表1

编号:□□□□ 患者姓名拼音缩写:□□□□ 病例报告表 (Case Report Form,CRF) 患者姓名拼音缩写:□□□□ 试验编号:□□□□ 医院名称: 研究者签名: 申办单位:

病例报告表填写要求 1、所有记入本手册的数据需对照原始资料进行核查 2、用签字笔填写,中文字应清晰可辨,英文字母需大写 3、每格填写一个字,不适用的空格,请填“/” 4、选择项请打“√” 5、填写错误修改时,用单线划掉,切毋涂抹,原字迹需清晰可辨,并签名和注明修改时间 6、患者的姓名不得出现在本病例报告表中,患者姓名拼音缩写四格需填满,两字姓名填写两字拼音前两个字母,三字姓名填写三字首字母及第三字二字母,四字姓名填写每一个字的首子母。举例:张红ZHHO,张红旗ZHQI 欧阳予黄OYYH。 7、日期填写为:□□□□年□□月□□日。

一、入选标准: 1、年龄18~65岁,性别不限;□是□否 2、创面面积小于体表面积的50%;□是□否 3、签署知情同意书;□是□否如以上任何答案为“否”,则该病人不能入选 二、排除标准: 1、年龄小于18岁或大于65岁;□是□否 2、有严重心、脑、肝、肾功能不全的患者;□是□否 3、严重营养不良的患者;□是□否 4、创面面积大于体表面积50%的患者;□是□否 5、有精神疾病的患者以及无自知力、不能确切表达者。□是□否 如以上任何答案为“是”,则该病人不能入选 观察医师签名:日期年月日

受试者一般资料 姓名:□□□□性别:男□女□年龄:□□岁 出生日期:□□□□年□□月□□日 体重:□□□㎏病例号:□□□□□□ 创面资料: 创面原因:□火焰□热液□化学□电烧伤□其他创面形成时间:□□月□□日创面部位: 创面面积:□□□□cm2肉芽:无□有□

临床试验数据管理系统-RealData!

临床试验数据管理系统-RealData!1 临床试验数据管理工作技术指南 2012年3月12日 目录 一、概述(1) 1.1国内临床试验数据管理现状(1) 1.2国际临床试验数据管理简介(2) 1.2.1ICH-GCP 对临床试验数据管理的原则性指导(3) 1.2.221 CFR Part 11对电子记录和电子签名的基本要求(4) 1.2.3“临床试验中采用计算机系统的指导原则”提供计算机系统开发的参照标准(4) 1.2.4GCDMP (Good Clinical Data Management Practice)提供全面具体的数据管理要求(5) 二、数据管理相关人员的责任、资质及培训(6) 2.1相关人员的责任(7) 2.1.1申办者(7) 2.1.2研究者(7) 2.1.3监查员(8)

2.1.4数据管理员(8) 2.1.5合同研究组织(CRO)(9) 2.2数据管理人员的资质及培训(10) 三、临床试验数据管理系统(11) 3.1临床试验数据管理系统的重要性(11) 3.2数据质量管理体系的建立和实施(12) 3.3临床试验数据管理系统的基本要求(13) 3.3.1系统可靠性(13) 3.3.2临床试验数据的可溯源性(Traceability) (14) 3.3.3数据管理系统的权限管理(Access Control)(15) 四、试验数据的标准化(16) 4.1临床数据标准化的现状与发展趋势(16) 4.2临床试验的数据标准化(17) 4.2.1CDISC和HL7 (18) 4.2.2医学术语标准(21) 4.2.3临床试验报告的统一标准(CONSORT)(26) 五、数据管理工作的主要内容(28)

临床试验总结报告的设计与撰写

一、临床试验方案设计 临床试验方案由研究者或申办者拟订,应符合GCP要求。研究者和申办者均应在已制定的临床试验方案上签名并签署日期。 临床试验设计的基本原则:①代表性:受试者样本符合总体规律;②重复:结果经得起重复验证;③随机:受试者随机分配入组;④对照与盲法:避免条件误差与主观因素。 试验方案的格式包括:①封页:包括题目、申办者和临床试验机构的名称与地址,拟订日期;②正文:GCP要求的23项;③封底:各参与的临床试验机构与主要研究者、申办者的名称与联系方式;④主要参考文献。 临床试验方案设计主要内容(GCP第四章第十七条)有以下23条: (一)试验题目: (二)试验目的,试验背景,临床前研究中有临床意义的发现和与该试验有关得临床试验结果、已知对人体的可能危险与受益,及试验药物存在人种差异的可能; (三)申办者的名称和地址,进行试验的场所,研究者的姓名、资格和地址; (四)试验设计的类型,随机化分组方法及设盲的水平; (五)受试者的入选标准,排除标准和剔除标准,选择受试者的步骤,受试者分配的方法; (六)根据统计学原理计算要达到试验预期目的所需的病例数; (七)试验用药品的剂型、剂量、给药途径、给药方法、给药次数、疗程和有关合并用药的规定,以及对包装和标签的说明; (八)拟进行临床和实验室检查的项目、测定的次数和药代动力学分析等; (九)试验用药品的登记与使用记录、递送、分发方式及储存条件; (十)临床观察、随访和保证受试者依从性的措施; (十一)中止临床试验的标准,结束临床试验的规定; (十二)疗效评定标准,包括评定参数的方法、观察时间、记录和分析; (十三)受试者的编码、随机数字表及病例报告表的保存手续; (十四)不良事件的记录要求和严重不良事件的报告方法、处理措施、随访的方式、时间和转归; (十五)试验用药品编码的建立和保存,揭盲方法和紧急情况下破盲的规定; (十六)统计分析计划,统计分析数据集的定义和选择; (十七)数据管理和数据可溯源性的规定; (十八)临床试验的质量控制和质量保证; (十九)试验相关的论理学; (二十)临床试验预期的进度和完成日期;

临床研究病例报告表

XXXXXXXXXXXXXXX XXXXXXXXXXXXXXXX XXXXXXXXXXXXX 临床研究病例报告表 (X日用药) 受试者姓名缩写□□□□ 药物编号:□□□ 试验中心编号:□ 试验开始日期年月日

填表说明 1.请用签字笔填写,字迹应清晰,易于辨认。 2.受试者姓名按汉语拼音首字母缩写靠左对齐填写。 3.在所有选择项目中,请在相应的方框中划“×”;疼痛强度(PI)和疼痛缓解度等数字表示项目请在选择的相应数字上画圈“○”。 4.每项填写内容务必准确,不得随便涂改,如发现填写内容有误,应在原记录上划单横线,在旁边写明正确内容,并注名修改者及日期。不要用任何方式(橡皮、涂改液等)涂抹原记录。 5.不要改变病例报告表的格式,如发现表中没有位置填写记录者希望记录的资料时,请将有关信息记录于后面的空白附页中,并保留以上记录副本。 6.知情同意书一般为患者签名。如患者有特殊情况,可由患者法定代理人签名。

试验观察流程图 入组筛选表

1.受试者应为: ?年龄:18 -70岁癌症患者(一般情况尚好,可适当放宽) ?性别不限 ?预计生存期2个月以上的住院患者 ?疼痛强度为中到重度,评分≥4 ?并符合一下条件之一(请在符合的项目上画圈) 1)入选前1周内曾使用XX,日剂量为40-60mg,疼痛强度可缓解到≤2; 2)入选前1周内曾使用其它镇痛药,其全日剂量相当于40-60mgXX剂量,疼痛强度可缓解到≤2 ?非放疗期或疼痛部位为非照射部位 ?间歇期的化疗者,应由主管医生确认化疗后无止痛作用 ?该患者是否同意参加本试验,并已签署知情同意书 如以上任何一个答案为“否”,此受试者不能参加 2.受试者排除标准: ?本研究开始前4周内曾参加过其他临床试验 ?正在服用或本试验开始前2周内曾服用MAO抑制剂者(如优降宁、苯乙肼等) ?24小时内用过XX类镇痛药,或5日内用过XXX ?癌痛骨转移患者,近4周内接受同位素内放疗或/和双磷酸盐类药物治疗 ?呼吸抑制、(肺)气道阻塞或组织缺氧 ?胆道疾病 ?心脏疾患(即Ⅱ级和Ⅱ级以上心功能) ?血压高于正常值 ?血液系统疾病 ?肝、肾功能明显异常(即指标高于正常值一倍以上) ?脑部疾病,判定能力异常 ?XX药耐受、过敏,或曾在使用时因不良反应停药 ?药物及∕或酒精滥用 ?孕妇或哺乳期妇女 如以上任何一个答案为“是”,此受试者不能参加 结论:该患者是否符合上述要求,同意入组是 □ □ □ □ □ □ □ □ 是 □ □ □ □ □ □ □ □ □ □ □ □ □ □ □ 否 □ □ □ □ □ □ □ □ 否 □ □ □ □ □ □ □ □ □ □ □ □ □ □ □ 医生签名:______日期____年___月___日 病历简况 1.1检查日期:年月日

药物临床试验总结报告的设计规范

药物临床试验总结报告的设计规范 版本号 1.0 页数11页 起草人起草日期年月日审核人审核日期年月日批准人批准日期年月日颁布日期年月日起效日期年月日 威海市立医院 药物临床试验机构

药物临床试验总结报告的设计规范 一目的 为真实地反应反映该试验的实施过程和客观地表述药物的有效性和安全性及其应用价值。 二范围 所有由本院牵头或协助参加的新药临床试验。 三内容 1报告封面 参照国家食品药品监督管理总局有关药品注册申报资料的形式要求。 2签名页 1)报告题目 2)执笔者签名。 3)主要研究者对研究试验报告的声明。 申明已阅读了该报告,确认该报告准确描述了试验过程和结果。 4)主要研究者签名和日期。(包括统计学工作者) 3报告目录 每个章节、附件、附表的页码。 4缩略语 正文中首次出现的缩略语应规范拼写,并在括号内注明中文全称。应以列表形式提供在报告中所使用的缩略语、特殊或不常用的术语定义或度量单位。 5伦理学问题 1)确认试验实施符合赫尔辛基宣言及伦理学原则。 2)伦理委员会组成及批准临床试验方案情况说明,并在附件中提供独立伦理委员会成 员表。 3)描述如何及何时获得与受试者入选相关的知情同意书,并在附件中提供原稿。 6报告摘要 报告摘要应当简洁、清晰的说明以下要点,通常不超过1500字。 1)试验题目。 2)临床批件文号。 3)主要研究者和临床试验单位。 4)试验的起止日期(第一例受试者第一次访视至最后一名受试者最后一次访视日期)。

5)试验目的及观察指标。 6)对研究药物的作用类别和主治功能的描述。 7)对试验设计作简短描述,包括试验设计类型(平行、交叉、成组序贯等)、设盲水 平(双盲、单盲或开放)、随机分组方法、对照的形式(安慰剂、阳性药对照、剂 量对照)、疗程。 8)试验人群。 9)给药方案(包括对照组) 10)评价标准(有效性和安全性评价指标)。 11)统计分析方法或模型(包括基线评价、组间比较、协变量分析、综合比较等)。 12)受试者入组情况及各组人口学资料。 13)各组疗效结果(主要和次要疗效指标)。 14)各组安全性结果(不良事件及严重不良事件)。 15)结论(有效性和安全性结论)。 7报告正文 1)前言 一般包括:受试药品研究背景;研究单位和研究者;目标适应症和受试人群、 治疗措施;受试者样本量;试验的起止日期;国家食品药品监督管理局批准临床试验的文号;制定试验方案时所遵循的原则、设计依据;申报者与临床研究单位之间有关特定试验的协议或会议等予以说明或描述。简要说明临床试验经过及结果。 2)试验目的 应提供对特定试验目的的陈述(包括主要、次要目的)。注意具体说明本项试验的受试因素、受试对象、研究效应,明确试验要回答的主要问题,明确药品的临床定位。 3)试验方法 (1)试验设计 a)总体研究设计和计划的描述(包括临床试验的流程图)。如试验过程中方案有 修正,应说明原因、更改内容及依据。 b)对试验总体设计的依据、合理性进行讨论,具体内容应视设计特点进行有针对 性的阐述。如采用单盲或开放试验设计,应说明理由。 c)提供样本含量的具体计算方法、计算过程以及计算过程中所用到的统计量的估 计值及其来源依据。 d)描述期中分析计划。

试验数据记录及修改的标准操作规程

试验数据记录及修改的标准操作规程 版本号 1.0 页数1页 起草人起草日期年月日审核人审核日期年月日批准人批准日期年月日颁布日期年月日起效日期年月日 威海市立医院 药物临床试验机构

试验数据记录及修改的标准操作规程 一、目的 规范试验数据记录标准操作规程,保证数据记录真实、及时、准确、完整,为临床试验的开展及总结或评价临床试验提供真实依据,保证药物临床试验质量。 二、范围 试验数据的记录过程。 三、内容 1.根据临床试验方案确定记录临床试验的信息,明确试验数据记录的要求。 2.对研究者、CRC、研究护士进行试验数据记录的培训。 3.及时、准确收集受试者的研究数据,反映受试者的病情变化、处理的过程及转归。 4.临床试验中各种实验室数据均应记录并保存,热敏纸类的检查单应予以复印。 5.实验室报告异常数值研究者及时进行有无临床意义,是否SAE/AE的判断,并签名。 6.记录及时、准确、完整,不随意删除、修改或增减数据,不伪造、编造数据。 7.试验数据如需修改,应将错误之处划线,不可涂黑,保证修改前记录能够辨认,在右上 角写上正确的内容、改正日期,并应有研究者签署姓名; 8.所有文字数据资料一律用蓝、黑钢笔,黑色水笔或签字笔书写,不得用铅笔、圆珠笔, 字迹清楚端正。 9.对显著偏离或在临床可接受范围以外的数据产生异议的,研究者做合理的解释,必要时 需进行复核。 10.客观记录受试者自觉症状,不诱导或暗示。按方案规定的随访时间和方法对试验客观指 标进行观察或检测。 11.核实所有观察结果和发现,以保证数据的可靠性。 四、参考资料 1.国家食品药品监督管理局令第3号发布《药物临床试验质量管理规范》,2003 第 1 页共1 页

药物临床试验基本流程(总结)

药物临床试验基本流程 1.临床试验启动阶段 1.1获得药物临床试验批件(有效期3年) 1.1制作研究者手册 理化性质、药理、药化、药效、毒理以及已有的临床研究资料,制作研究者手册。 研究者手册是临床试验开始前的资料汇编。研究者手册的内容一般包括:目录、序言、化学和物理性质、临床前研究、药理学研究、药代动力学研究、毒理学研究、已有临床资料、药品使用信息。 1.2筛选主要研究者 CFDA 网站中浏览临床药理基地名录,筛选医院(一般来说不是整个医院而是 某个科室),然后选择合适的主任级医生; 联系主任医生(看是否有意向,在给予任何资料前,签保密协 议,然后发放可行性问卷,收集可行性问卷,确认完整无误,回复感谢信, 感谢其参与问卷调查) 如果可行,可安排访视,具体考察该基地是否有能力执行临床试验; (电话预约研究者,给予方案和研究者手册,agenda 确认要讨论的具体问题) Agenda:研究人员的分配 方案的可行性 能否入够病人及如何保证其依从性 药品管理 档案管理 研究者职责 主要研究者资质的确认 相关研究人员的资质 硬件设施考察 伦理委员会的访视(承不承认中心伦理、伦理开会频率、送审清单、伦理委员会人员清单) 书写访视报告; 再次拜访,与其讨论方案、试验时间、试验费用等问题; 1.3试验文件准备 会同CRO 、申办者、主要研究者,共同制定临床试验方案、病例报告表、知情同意书样本、原始文件;CRO 制定临床试验中其他用表; 1.4其他研究者筛选 从临床基地名录中选择其他可能的研究者,可根据首研者的推荐以及公司曾经合作的情况进行综合判断; 准备资料:方案、研究者手册、知情同意书样本、病例报告表; 与其谈论方案,要求提供其医院所能提供的病例数、时间进度和经费预算; 选定合适研究者,征得其医院管理部门的同意; 1.5召开多中心临床方案认论会(研究者会议),讨论试验方案、CRF 等 注:是否需要召开研究者会议要看具体情况 1.6取得伦理委员会批件 注:IRB (Independent Review Board )/IEC (Independent Ethics Committee )——IRB 为美国的伦理委员会,IEC/EC 为欧盟的伦理委员会 按照GCP 的要求,所有临床试验必须得到伦理委员会的批准。在实际进行临床试验时,首先必须取得牵头单位伦理委员会的批准,其他参加单位是否要过伦理根据各家单位的具体要求而定。 伦理委员会将对有关批件、药检报告、研究者手册、知情同意书样本、试验方案(protocal )、病例报告表进行审批; 1.7试验药品准备 督促申办方进行试验用药品的送检; 生物统计师设计随机分组方案; 根据随机分组方案,设计药品标签; 设计应急信件; 药品包装:为每一个受试者准备好一盒药物,药盒上贴好标签,并装入相应的应 先拜访研究者还是医院相关部门视具体情况而定,没有统一标准 1.7及1.8可在研究者会议后开始准备,同时申请伦理委员会批件

病例报告表记录标准操作规程

病例报告表记录标准操作规程 1.目的:为加强对药物研究的监督管理,保证药物临床病历报告表 记录真实、规范、完整,提高药物临床试验的质量,根据《中华人民共和国药品管理法》、《国家档案法》以及药物申报和审批中的有关要求,制订本规程。 2.凡在我机构进行的药物临床试验,其病例报告表记录均应遵循本 操作规程。 3.药物临床试验病例报告表是指按试验方案所规定设计的一种文 件,用以记录每一名受试者在试验过程中的数据。每位受试者在试验中的有关资料均应记录于病例报告表中。 4.病例报告表记录的基本要求:真实、及时、准确、完整、规范, 防止漏记和随意涂改。不得伪造、编造数据。为确保达到以上要求,记录时需注意以下事项: 4.1 病例报告表中的数据来自原始文件并与原始文件一致,试验中 的任何观察、检查结果均应及时、准确、完整、规范、真实地记录于病历和正确地填写至病例报告表中,不得随意更改,确因填写错误,作任何更正时应保持原始记录清晰可辨,由更正者签署姓名和时间; 4.2 临床试验中各种实验室数据均应记录或将原始报告复印件粘贴 在病例报告表上,在正常范围内的数据也应具体记录。对显著偏离或在临床可接受范围外的数据须加以核实。检测项目必须注明所采用的计量单位; 4.3 病例报告表必须使用本研究机构统一专用的带有页码编号的病 例报告表。病例报告表的幅面,根据需要设定。 4.4 病例报告表中记录的内容要有必要的依据。计算机、自动记录 仪器打印的图表和数据资料,临床研究中的检验报告书、体检表、知情同意书、试验图片、照片等应按顺序粘贴在病例报告表的相应位置上,并在相应处注明试验日期和时间;底片装在统一制作的底片袋内,编号后另行保存。用热敏纸打印的试验记录,须保留其复印件。不宜粘贴的,可另行整理装订成册并加以编号,同时在病例报告表相应处注明,以便查对。 4.5 病例报告表应保持完整,不得缺页或挖补;如有缺页、漏页, 应详细说明原因。 4.6 病例报告表的记录不得使用容易擦改的记录工具如铅笔等,宜

医疗器械临床试验病例报告表范本

附件3 医疗器械临床试验病例报告表范本试验项目名称:受试者病例号:试验用医疗器械编号:临床试验机构(或编号):临床试验 方案编号:临床试验方案版本号和日期:试验开始日期:年 月日随访结束日期:年月日记录人签名: 1 填表说明: 1. 每项内容填写应当准确、完整、及时、可读。 2. 记录的试验数据和信息应当与原始文件一致。 3. 填写内容不得随意更改,如发现内容有误,应当在原记录上划单横线,不得用任何方式掩盖,并在旁边注明正确内容及修改原因,由研究者签名并注明日期。 2 一、受试者病历简况 1.签署知情同意日期年月日入组日期年月日2.受试者姓名缩写及鉴认代码 3.出生日期年月 4.性别男□ 女□ 5.临床诊断 6.接受的诊断/治疗方式(既往病史、用药和/或手术) 7.入组前基础症状 8.入组时体格检查、实验室检查二、符合入选/排除标准三、诊断/治疗情况记录四、随访情况记录五、一般情况观察记录(伴随疾病/药物)六、不良事件记录表如果在试验期间没

有不良事件发生,请在此□中打“×”,并在此表下方签名。如有请用标准医学术语记录所有观察到的不良事件(包括直接询问出的)。每一栏记录一个不良事件。不良事件描述开始发生时间1结束时间不良事件□阵发性发作次数□□特点□持续性 3 □ 轻不良事件记录□ 中2报告及程度□ 重报告有□ 无□ 严重不良事件□是□否□ 肯定有关□ 很可能有关与试验□ 可能有关的关系□ 可能无关□ 无关□消失后遗症有□ 无□ 转归□继续□死亡□是□否纠正治疗因不良事件而□是□否退出试验 1. 如果不良事件仍存在,请不要填写此项。 2. 程度: 症状按轻(询问出);中(主动叙述但能忍耐);重(有客观备注表现,难忍耐)填写。七、严重不良事件和器械缺陷记录八、试验方案的偏离情况记录九、试验完成情况十、声明此病例报告表中的信息记录真实、准确,符合试验方案的要求,特此声明。研究者签名:年月日 4

临床试验总结报告的体例格式和内容要求

临床试验总结报告体例和内容要求 1.题目封面 封面题页应包括如下内容: ——试验题目 ——试验药物/研究产品的名称 ——试验用药品的适应症 ——如不能在题目中表明、则简要描述(以1~2句话)设计(平行、交叉、双盲、随机)比较(安慰剂、活性成分、剂量/反应),间隔,剂量和病例数。 ——申办者姓名 ——原始规划与计划的确认(编码或数目,确认日期) ——试验预期进度与进程安排 ——研究开始数据(受试的第一位病人,或任何其它数据) ——后期研究终末数据(最终一个受试者的名称、或终末研究者完成的数据) ——试验研究完整的数据(最后的受试者) ——参加者或合作者或主办者责任医务官等人士的名称和职责。 ——药业公司名称/主办方签名(在公司/主办者中负责研究报告的人,公司/主办方联络者的姓名,电话和传真号码,以备在回顾研究报告提出问题时,在此页码或应用的字母有所表明)。 ——表明该项研究是否在优良临床试验管理规范(GCP)条件下进行的包括应达到的基本文件、研究设施、人员知识结构,统一培训等。 ——报告的资料(通过题目和资料,确定与该项研究相同的任何其它较早报告) 2.内容概述 应提供—个临床试验的主要内容提要(通常限于3页内容),该提要应包括试验题目、研究人员、研究出版物名称、试验时间、试验目的、试验方法、试验研究样本、诊断及进入研究的主要标准、试验的产品、剂量、给药方式及批号、治疗持续时间、参考的治疗、剂量、给药方式及批号、评价标准(有效性、安全性)、统计方法、总结——结论(效能结论、安全性结论、结论)及报告日期。该提要应包括表明结果的数字资料,而不仅仅是文字和P 值(见附件l,研究内容概述)。 3.内容目录 内容表格应包括: ——页码或其它每一个部分的局部资料,包括归纳性表格、图和图表。 ——附录、列表和任何能提供的病例报告形式的汇总和定位。 4.名词、术语缩写和定义 应提供报告中使用的—系列缩写符、特殊的或不常用的术语,或测量单位定义,应拼写

临床试验数据管理工作技术指南

附件 临床试验数据管理工作技术指南 一、概述 临床试验数据质量是评价临床试验结果的基础。为了确保临床试验结果的准确可靠、科学可信,国际社会和世界各国都纷纷出台了一系列的法规、规定和指导原则,用以规范临床试验数据管理的整个流程。同时,现代新药临床试验的发展和科学技术的不断进步,特别是计算机、网络的发展又为临床试验及其数据管理的规范化提供了新的技术支持,也推动了各国政府和国际社会积极探索临床试验及数据管理新的规范化模式。 (一)国内临床试验数据管理现状 我国的《药物临床试验质量管理规范》(Good Clinical Practice,GCP)对临床试验数据管理提出了一些原则要求,但关于具体的数据管理操作的法规和技术规定目前还处于空白。由于缺乏配套的技术指导原则,我国在药物临床试验数据管理方面的规范化程度不高,临床试验数据管理质量良莠不齐,进而影响到新药有效性和安全性的客观科学评价。此外,国内临床试验中电子化数据管理系统的开发和应用尚处于起步阶段,临床试验的数据管理模式大多基于纸质病例报告表(Case Report Form,CRF)的数据采集阶段,电子化数据采集与数据管理系统应用有待推广和普及。同时,由于缺乏国家数据标准,同类研究的数据库之间难以做到信息共享。

(二)国际临床试验数据管理简介 国际上,人用药品注册技术要求国际协调会议的药物临床研究质量管理规范(以下简称ICH E6 GCP)对临床试验数据管理有着原则性要求。对开展临床试验的研究者、研制厂商的职责以及有关试验过程的记录、源数据、数据核查等都直接或间接地提出了原则性的规定,以保证临床试验中获得的各类数据信息真实、准确、完整和可靠。 各国也颁布了相应的法规和指导原则,为临床试验数据管理的标准化和规范化提供具体的依据和指导。如:美国21号联邦法规第11部分(21 CFR Part 11)对临床试验数据的电子记录和电子签名的规定(1997年),使得电子记录、电子签名与传统的手写记录与手写签名具有同等的法律效力,从而使得美国食品药品管理局(FDA)能够接受电子化临床研究材料。据此,美国FDA 于2003年8月发布了相应的技术指导原则,对Part 11的规定作了具体阐释,并在计算机系统的验证、稽查轨迹,以及文件记录的复制等方面提出明确的要求。 2007年5月,美国FDA颁布的《临床试验中使用的计算机化系统的指导原则》(Guidance for Industry: Computerized Systems Used in Clinical Investigations)为临床试验中计算机系统的开发和使用提供了基本的参照标准。 而且由国际上相关领域专家组成的临床试验数据管理学会(Society of Clinical Data Management, SCDM)还形成了一部《良好的临床数据管理规范》(Good Clinical Data Management Practice,GCDMP),该文件为临床试验数据管理工作的每个关键环节都规

临床试验数据核查指导原则

附件 3 仿制药质量和疗效一致性评价 临床试验数据核查指导原则 为贯彻落实《国务院办公厅关于开展仿制药质量和疗效一致性评价的意见》(国办发〔2016〕8号),进一步规范仿制药质量和疗效一致性评价(以下简称一致性评价)临床试验数据核查要求,保证药品检查质量,制定本指导原则。 式审查。一般在形式审查后30日内组织临床试验数据核查。 2. 省级食品药品监督管理部门结合申请人提交的一致性评价申报资料、《仿制药质量和疗效一致性评价临床试验数据核查申报表》(附1)制定核查方案,选派核查组。核查组一般由2~4名核查员组成,核查组按照核查方案开展核查,并完成《仿制药质量和疗效一致性评价临床试验数据现场核查发现问题》(附2)、《仿制药质量和疗效一致性评价临床试验数据现场核查报告》(附3)。 3. 省级食品药品监督管理部门对《仿制药质量和疗效一致性评价临床试验数据现场核

查报告》进行审核。 4.核查中心依据各省一致性评价临床试验现场核查进展情况,定期制定抽查计划,开展监督检查工作。 (二)进口仿制药品 1.国家食品药品监督管理总局行政事项受理服务和投诉举报中心对一致性评价资料和补充申请资料进行接收、受理和形式审查,形式审查后将资料送核查中心。 2. 对申请人提交的进口仿制药品的国内临床研究数据,核查中心一般在收到资料后30 (三)确保数据的真实性、可靠性和临床试验开展的合规性 1.真实性 开展一致性评价工作应当坚持诚实守信,确保申报资料与原始记录的真实性,禁止任何虚假行为。 2.可靠性 应确保申报资料与原始数据一致。应当规范一致性评价过程中的记录与数据的管理,保证数据记录准确真实、清晰可追溯、原始一致、及时同步记录、能归属到人、完整持久,

(完整)医疗器械临床试验病例报告表

(完整)医疗器械临床试验病例报告表 编辑整理: 尊敬的读者朋友们: 这里是精品文档编辑中心,本文档内容是由我和我的同事精心编辑整理后发布的,发布之前我们对文中内容进行仔细校对,但是难免会有疏漏的地方,但是任然希望((完整)医疗器械临床试验病例报告表)的内容能够给您的工作和学习带来便利。同时也真诚的希望收到您的建议和反馈,这将是我们进步的源泉,前进的动力。 本文可编辑可修改,如果觉得对您有帮助请收藏以便随时查阅,最后祝您生活愉快业绩进步,以下为(完整)医疗器械临床试验病例报告表的全部内容。

编号:□□□□ 患者姓名拼音缩写:□□□□ 产品名称*****(商品名:***) 的安全性和有效性研究 病例报告表 (Case Report Form,CRF) 患者姓名拼音缩写:□□□□ 试验编号:□□□□ 医院名称: 研究者签名: 申办单位:上海******有限公司

病例报告表填写要求 1、所有记入本手册的数据需对照原始资料进行核查 2、用签字笔填写,中文字应清晰可辨,英文字母需大写 3、每格填写一个字,不适用的空格,请填“/" 4、选择项请打“√" 5、填写错误修改时,用单线划掉,切毋涂抹,原字迹需清晰可辨,并签名和注明修改时间 6、患者的姓名不得出现在本病例报告表中,患者姓名拼音缩写四格需填满,两字姓名填写两字拼音前两个字母,三字姓名填写三字首字母及第三字二字母,四字姓名填写每一个字的首子母。举例:张红ZHHO,张红旗ZHQI 欧阳予黄 OYYH. 7、日期填写为:□□□□年□□月□□日。

一、入选标准: 1、年龄18~65岁,性别不限;□是□否 2、创面面积小于体表面积的50%;□是□否 3、签署知情同意书; □是□否 如以上任何答案为“否”,则该病人不能入选 二、排除标准: 1、年龄小于18岁或大于65岁;□是□否 2、有严重心、脑、肝、肾功能不全的患者;□是□否 3、严重营养不良的患者;□是□否 4、创面面积大于体表面积50%的患者;□是□否 5、有精神疾病的患者以及无自知力、不能确切表达者。□是□否 如以上任何答案为“是",则该病人不能入选 观察医师签名: 日期年月日

临床试验观察表(CRF)29652

病例报告表(Case Report Form) 受试者姓名: 家庭地址: 联系电话: 试验中心名称: 申办单位:延吉喜来健医疗器械有限公司

在正式填表前请认真阅读下列填表说明 病例报告表填写说明: 1.筛选合格者填写病例报告表。 2.病例报告表填写务必准确、清晰,不得任意涂改,错误之处纠正时需用横线居中化出,并签署修改者姓名缩写及修改时间。 3.填写记录一律用钢笔或碳素签字笔。 4.患者姓名拼音缩写四个需填满,两字姓名填写两字拼音前两个字母;三字姓名填写三字首字母及第三字第二字母;四字姓名填写每个字的首字母。5.表中凡有“□”的项,请在符合的条目上划“√”。表格中所有栏目均应填写相应的文字或数字,不得留空。 6.所有检验项目因故未查或漏查,请填写ND;具体合并用药剂量和时间不明,请先写NK。 7.试验期间应如实填写合并用药记录表、不良事件记录表。记录不良事件的发生时间、严重程度、持续时间、采取的措施和转归。如有严重不良事件发生(包括临床验证过程中发生需住院治疗、延长住院时间、伤残、影响工作能力危及生命或死亡、导致先天畸形等事件),必须立即通知主要研究单位伦理委员会及申办单位。

8.临床试验应严格按照临床试验方案要求进行。试验不同时期需完成的检查和需记录的项目,请对照临床流程图执行。

受试者知情同意书 敬爱的患者: 我们现在正在进行一项临床研究,该项临床研究是经吉林省药品监督管理局备案的,研究的目的是评价物理治疗的高科技产品----温热电位治疗仪的疗效和安全性。 温热磁疗仪是XXX医疗器械有限公司根据韩国专利技术研制生产的医疗器械,本治疗仪是通过XXXXXXXXXXXXXXXXXXXXXXXXXXXXXX机理,主要用于XXXXXXXXXXXXX 治疗。具有平衡机体阴阳,增强脏腑机能,促进新陈代谢,调节植物神经,改善心脑血管血液供应,促进血液循环,调节血管张力,降低血液粘稠度,防治动脉粥样硬化,促进组织的

临床研究病例报告表三篇

临床研究病例报告表三篇 篇一:临床研究病例报告表 受试者姓名缩写□□□□ 药物编号:□□□ 试验中心编号:□ 试验开始日期年月日 填表说明 1.请用签字笔填写,字迹应清晰,易于辨认。 2.受试者姓名按汉语拼音首字母缩写靠左对齐填写。 3.在所有选择项目中,请在相应的方框中划“×”;疼痛强度(PI)和疼痛缓解度等数字表示项目请在选择的相应数字上画圈“○”。4.每项填写内容务必准确,不得随便涂改,如发现填写内容有误,应在原记录上划单横线,在旁边写明正确内容,并注名修改者及日期。不要用任何方式(橡皮、涂改液等)涂抹原记录。 5.不要改变病例报告表的格式,如发现表中没有位置填写记录者希望记录的资料时,请将有关信息记录于后面的空白附页中,并保留

以上记录副本。 知情同意书一般为患者签名。如患者有特殊情况,可由患者法定代理人签名。 试验观察流程图

入组筛选表

1.受试者应为: ?年龄:18-70岁癌症患者(一般情况尚好,可适当放宽) ?性别不限 ?预计生存期2个月以上的住院患者 ?疼痛强度为中到重度,评分≥4 ?并符合一下条件之一(请在符合的项目上画圈) 1)入选前1周内曾使用XX,日剂量为40-60mg,疼痛强度可缓解到≤2; 2)入选前1周内曾使用其它镇痛药,其全日剂量相当于 40-60mgXX剂量,疼痛强度可缓解到≤2 ?非放疗期或疼痛部位为非照射部位 ?间歇期的化疗者,应由主管医生确认化疗后无止痛作用 ?该患者是否同意参加本试验,并已签署知情同意书 如以上任何一个答案为“否”,此受试者不能参加 2.受试者排除标准: ?本研究开始前4周内曾参加过其他临床试验 ?正在服用或本试验开始前2周内曾服用MAO抑制剂者(如优降宁、苯乙肼等) ?24小时内用过XX类镇痛药,或5日内用过XXX 是 □ □ □ □ □ □ □ □ 是 □ □ □ □ □ □ □ □ □ 否 □ □ □ □ □ □ □ □ 否 □ □ □ □ □ □ □ □ □

临床试验—病例表

甘露聚糖肽注射液辅助治疗恶性肿瘤的安全性和有效性的随机、双盲、与安慰剂平行对照、多中心临床再评价试验 病例报告表 (Case Report Form) 受试者姓名缩写□□□□ 药物编号:□□□ 试验中心编号:□□ 试验开始日期年月日

填表说明 1.请用签字笔填写,字迹应清晰,易于辨认。 2.受试者姓名按汉语拼音首字母缩写靠左对齐填写。 3.在所有选择项目中,请在相应的方框中划“×”;疼痛强度(PI)和疼痛缓解度等数字表示项目请在选择的相应数字上画圈“○”。 4.每项填写内容务必准确,不得随便涂改,如发现填写内容有误,应在原记录上划单横线,在旁边写明正确内容,并注名修改者及日期。不要用任何方式(橡皮、涂改液等)涂抹原记录。 5.不要改变病例报告表的格式,如发现表中没有位置填写记录者希望记录的资料时,请将有关信息记录于后面的空白附页中,并保留以上记录副本。 6.知情同意书一般为患者签名。如患者有特殊情况,可由患者法定代理人签名。

试验观察流程图

入组筛选表 1.受试者应为: ?年龄:18 -70岁癌症患者(一般情况尚好,可适当放宽) ?性别不限 ?预计生存期2个月以上的住院患者 ?疼痛强度为中到重度,评分≥4 ?并符合一下条件之一(请在符合的项目上画圈) 1)入选前1周内曾使用XX,日剂量为40-60mg,疼痛强度可缓解到≤2; 2)入选前1周内曾使用其它镇痛药,其全日剂量相当于40-60mgXX剂量,疼痛强度可缓解到≤2 ?非放疗期或疼痛部位为非照射部位 ?间歇期的化疗者,应由主管医生确认化疗后无止痛作用 ?该患者是否同意参加本试验,并已签署知情同意书 如以上任何一个答案为“否”,此受试者不能参加 2.受试者排除标准: ?本研究开始前4周内曾参加过其他临床试验 ?正在服用或本试验开始前2周内曾服用MAO抑制剂者(如优降宁、苯乙肼等) ?24小时内用过XX类镇痛药,或5日内用过XXX ?癌痛骨转移患者,近4周内接受同位素内放疗或/和双磷酸盐类药物治疗 ?呼吸抑制、(肺)气道阻塞或组织缺氧 ?胆道疾病 ?心脏疾患(即Ⅱ级和Ⅱ级以上心功能) ?血压高于正常值 ?血液系统疾病 ?肝、肾功能明显异常(即指标高于正常值一倍以上) ?脑部疾病,判定能力异常 ?XX药耐受、过敏,或曾在使用时因不良反应停药 ?药物及∕或酒精滥用 ?孕妇或哺乳期妇女 如以上任何一个答案为“是”,此受试者不能参加 结论:该患者是否符合上述要求,同意入组是 □ □ □ □ □ □ □ □ 是 □ □ □ □ □ □ □ □ □ □ □ □ □ □ □ 否 □ □ □ □ □ □ □ □ 否 □ □ □ □ □ □ □ □ □ □ □ □ □ □ □ 医生签名:______日期____年___月___日 病历简况

病例报告表(样板)

碳水化合物交换份法在糖尿病饮食干预中的应用研究多中心随机对照临床试验 病例报告表 (Case Report Form,CRF) □ 01 医院 □ 02 医院 □ 03 医院 □04 医院 受试者姓名(拼音缩写):□□□□ 研究者签名: 申办者:浙江医院

注意事项 1.本试验主要研究者必须经过GCP培训,必须事先充分了解试验方案及有关资料,严格按方案执行,病例筛选合格者按数字随机表分组,不得随意更改分配。 2.筛选合格者填写正式病例报告表,病例报告表填写务必准确、清晰,不得随意涂改,错误之处纠正时需用双横线居中划出,并签署修改者姓名缩写及修改时间。 3.每页页眉均应填写,其中中心代码填 1,2,3,4等;入组顺序号由各医院按受试者就诊时间顺序填写(入组顺序号的次序应与就诊先后时间及住院号顺序相符合); 随机对应号请按试验设计要求填写;受试者姓名拼音缩写四格应填满,两字姓名按每个字前两个字母填写;三字姓名按每个字的首个字母和第三个字的第二个字母填写;四字姓名按每个字的首个字母填写。举例:张红 ZHHO 李书名 LSMI 欧阳小惠 OYXH 4.血糖因故未查或漏查,应填写ND。 5.填写数字时应将□都填满,位数不够的靠右侧填写,左侧空出填“0”。如□0□4。

入组选择表 请回答以下问题 纳入标准是否无关1. 2014年6月-2016年6月收治内分泌科被诊断为2型 □□□糖尿病患者; 2. 年龄在18~80岁之间;□□□ 3. 确诊2型糖尿病1年以上□□□ 4. 受试者自愿参加研究并己签署知情同意书□□□ 5. 神志清,可自行进行饮食控制;□□□ 上述问题任一回答是“否”时,该病例不能纳入研究 排除标准是否无关 1. 预计住院时间不满3天□□□ 2. 近期接受过大手术或任何一项超过2小时的手术□□□ 3. 正在接受糖皮质激素治疗者□□□ 4. 正在接受肠内或肠外营养□□□ 5. 肝、肾功能障碍及其他禁忌症□□□ 6. 糖尿病酮症酸中毒□□□ 7. 妊娠期女性□□□ 8. 不愿意接受饮食管理或无法签署知情同意书者□□□ 上述问题任一回答是“是”时,该病例不能纳入研究 研究者签名日期□□□□年□□月□□日

I期临床试验总结报告样本

SFDA临床试验批准号:2002XXXXXX号 XXX注射液 Ⅰ期临床人体耐受性试验总结报告 试验单位:XXXXXXXX医院 试验负责:XXX 试验设计:XXX,XXXX 试验日期:2003年X月X日至X月X日 申办单位:XXXXX有限公司 (以下仅提供报告格式,其数据和文字均为虚构,如有巧合,纯属偶然;数据前后有矛盾之处,也请原谅) XXX注射Ⅰ期临床人体耐受性试验总结报告 摘要 单次给药的耐受性试验:30名健康受试者,根据体重和性别随机分配到7个剂量组(组1、组2、组3、组4、组5、组6、组7)。组1、组2每组2人;组3、组4、组5每组6人;组6、组7每组4人。参照费氏递增法(改良Fibonacci法)递增给药,各组分别静脉滴注XXX注射液5、10、20、30、40、50、60ml,滴速20-30滴/min。每个受试者只接受一个相应的剂量,从小剂量开始,每个剂量应用后未见不良反应,才可用下

一个剂量。观察给药后健康人体对XXX注射液的反应和耐受性。 多次(累积)给药的耐受性试验:12名健康受试者根据体重和性别随机分配到甲、乙两个剂量组,每组6人。分别静脉滴注XXX注射液20、10 ml,滴速20-30滴/min,连续5d。 结论:(1)每次按10 ml/次给药,有可能引血胆红素轻度增高或尿胆原阳性,但受试者无明显不适,因此推荐II期临床试验为10 ml/次;15 ml/次有可能引起血胆红素轻度增高或尿胆原阳性,但风险不会太大,可在严密监护下尝试。20 ml/次虽然引起的不良反应尚属轻度,但发生的频度较高,慎用。(2)10 ml/次就有可能引起轻度不良反应,主要为血胆红素轻度增高或尿胆原阳性,但受试者无自觉症状。20 ml/次可引起的不良反应有所增加。这种血胆红素轻度增高或尿胆原阳性,且两次重复结果相近,推测是药物引起的轻度溶血所致。其他主要不良反应为口干(2例)、头晕(1例)、皮疹(1例)。 根据国家药品监督管理局XXX号批文的要求,按照《新药审批办法》,《药品临床试验管理规范》(GCP),《中药新药临床研究的技术要求》,以及《中药新药临床研究指导原则》和XXX的化学组成、功能主治、药效学、毒理学研究资料,于2003年7月17日~2003年9月20日,对XXX有限公司申报的XXX注射液中药二类新药,进行Ⅰ期临床人体耐受性试验,总结报告如下。 1 研究目的 选择健康人为受试者,从安全的初始剂量开始,观察人体对XXX注射液的耐受性,为制定本品的Ⅱ期临床试验给药方案提供依据。 2 临床资料与研究方法 病例选择 入选标准 1.健康志愿者。 2.年龄在18~50岁,男女各半。 3.体重在标准体重的±10%范围内[标准体重(kg)=×(身高cm-80)]。 4.无烟酒嗜好。 5.心、肝、肾、血液等检查指标均在正常范围。 6.知情同意,志愿受试。 排除标准 1.妊娠期、哺乳期妇女。 2.具有原发性心、肝、肾、血液学疾病者。 3.精神或法律上的残疾患者。 4.有酒精、药物滥用病史。 5.过敏体质(对两种或两种以上药物或食物、花粉过敏史者)。 6.3个月内参加过其他药物临床试验者。 7.正在应用其他预防和治疗的药物者。 8.研究者认为不能入组的其他原因。 剔除标准 对已被选入本临床研究,属于以下情况之一者,作为剔除病例。 1.不符合纳入标准,或符合排除标准者

相关文档
最新文档