Clinical Trials - Study Design, Endpoints and Biomarkers, Drug Safety, and FDA and ICH Guidelines

Clinical Trials - Study Design, Endpoints and Biomarkers, Drug Safety, and FDA and ICH Guidelines

von: Tom Brody

Elsevier Reference Monographs, 2011

ISBN: 9780123919137 , 638 Seiten

Format: PDF, ePUB, OL

Kopierschutz: DRM

Windows PC,Mac OSX geeignet für alle DRM-fähigen eReader Apple iPad, Android Tablet PC's Apple iPod touch, iPhone und Android Smartphones Online-Lesen für: Windows PC,Mac OSX,Linux

Preis: 60,95 EUR

Mehr zum Inhalt

Clinical Trials - Study Design, Endpoints and Biomarkers, Drug Safety, and FDA and ICH Guidelines


 

Front Cover

1

Clinical Trials: Study Design, Endpoints and Biomarkers, Drug Safety, FDA and ICH Guidelines

4

Copyright Page

5

Contents

8

Acknowledgments

16

Preface

18

The Study Schema and Study Design

18

Intent to Treat Analysis

18

How to Choose the Endpoints

18

Diagnostic Tests

19

Mechanism of Action

19

Standards

19

Methodology

19

Clinicaltrials.Gov and other Registries for Clinical Trials

20

Introduction

24

Abbreviations and Definitions

28

Biography

34

1 The Origins of Drugs

36

I. Introduction

36

II. Structures of Drugs

37

a. Origins of warfarin

37

b. Origins of methotrexate and 5-fluorouracil

38

c. Origins of ribavirin

39

d. Origins of paclitaxel

39

e. Origins of cladribine

40

f. Origins of drugs in high-throughput screening

42

g. Origins of therapeutic antibodies

42

III. The 20 Classical Amino Acids

44

IV. Animal Models

46

a. Introduction

46

b. Estimating human dose from animal studies

48

1. NOAEL approach

49

2. MABEL approach

49

c. Scaling up the drug dose, acquired from animal studies, for use in humans

49

2 Introduction to Regulated Clinical Trials

52

I. Introduction

52

II. Study Design

54

III. The Study Schema

55

a. Examples of schema from clinical trials

57

b. Sequential treatment versus concurrent treatment – the Perez schema

59

c. Neoadjuvant chemotherapy versus adjuvant chemotherapy – the Gianni schema

61

d. Neoadjuvant chemotherapy plus adjuvant chemotherapy – the Untch schema

61

e. Forwards sequence and reverse sequence – the Puhalla schema

62

f. Both arms received three drugs, each arm at a different schedule – the Sekine schema

63

g. Staging – the Blumenschein schema

63

h. Staging and restaging – the Czito schema

65

i. Methodology tip – staging

65

j. Decision tree – the Baselga schema

66

k. Decision tree – the Katsumata schema

66

l. Methodology tip – what is “tumor progression”?

70

m. Methodology tip – unit of drug dose expressed in terms of body surface area

70

n. Run-in period – the schema of Dy

71

o. Methodology tip – c-kit and imatinib

72

p. Run-in period – the Hanna schema

72

q. How to maintain blinding of the treatment, when the study drug and the control treatment are provided by different-sized pills (or by different volumes of solutions)…

73

r. Methodology tip – bevacizumab and VEGF

76

s. Dose escalation – the Moore schema

76

t. Pharmacokinetics – the Marshall schema

78

IV. Further Concepts In Study Design

79

a. Active control

79

b. Add-on design active control

81

c. Three-arm study – clinical trial with two different active control arms

82

V. Summary

82

VI. Amendments to the Clinical Study Protocol

83

3 Run-In Period

86

I. Introduction

86

a. Washout period

87

b. Detecting baseline adverse events

87

c. Excluding potential study subjects who have safety issues correlating with the study drug

87

d. To include only study subjects with controllable pain

88

e. To determine the maximal tolerable dose

89

f. To achieve and ensure steady state in vivo concentrations of study drug

89

g. To allow a period of adjustment of lifestyle of the study subjects, for example changes in dietary patterns

90

h. To ensure that metabolic characteristics of all study subjects are similar, prior to administering drugs

91

i. To ensure that potential study subjects can adhere to, or comply with, the study protocol

91

j. To confirm that all study subjects meet the inclusion and exclusion criteria

92

k. Detecting potential study subjects who show a predetermined desired response to the study drug, with the goal of including only these subjects

93

l. Methodology tip – anti-cancer drugs that inhibit tumor growth

94

m. Decision tree

94

n. To create a self-control group

95

II. Concluding Remarks

96

4 Inclusion/Exclusion Criteria, Stratification, and Subgroups – Part I

98

I. The Clinical Study Protocol Is a Manual that Provides the Study Design

98

a. Clinical study protocol provides the inclusion/exclusion criteria and stratification

99

b. Stratification of study subjects

100

c. Words of warning

102

d. Staging of the disease

103

e. The study schema

103

1. Inclusion criteria for the RTOG 0232 study of prostate cancer

105

2. Exclusion criteria for the RTOG 0232 study of prostate cancer

105

f. Stratification of subjects into subgroups

105

g. Examples of subgroups

105

h. Prior therapy

107

i. Poor performance status as a basis for exclusion

108

j. Irreversible and cumulative toxicity as a basis for exclusion

109

k. Drug resistance as a basis for exclusion

111

II. Biology of Drug Resistance

111

a. Biochemistry of the ABC drug transporters

111

b. Biology of cross-resistance

112

c. A tumor’s genetic expression can provide guidance on drug resistance

113

1. Doxorubicin

113

2. Paclitaxel

113

3. Tamoxifen

114

4. Imatinib

114

d. Prior treatment with hormones as a basis for exclusion

115

e. Immune status for exclusion criteria

116

f. Example of earlier vaccination as an inclusion criterion

116

g. Ethical considerations as a basis for inclusion criteria

117

III. More Information on Subgroups

117

a. Subgroup of non-elderly subjects and subgroup of elderly subjects

118

b. Subgroup of subjects with no metastasis and subgroup of subjects with metastasis

119

c. Subgroup of smokers and subgroup of non-smokers

120

d. Subgroups set forth in a clinical study protocol can be used as a basis for FDA approval

120

e. Subgroup analysis enables recommendations for a specific course of treatment

121

f. Subgroup analysis can justify increases in drug dose for specific subgroups

122

g. Subgroups determined by an analysis of gene expression by microarray analysis

122

h. Recommending dropping one subgroup from the trial, rather than stopping the entire trial

123

IV. Concluding Remarks

124

5 Inclusion and Stratification Criteria – Part II

126

I. Introduction

126

II. Staging

126

a. History of tumor staging

127

b. Revising staging systems

127

c. Biology of tumors

128

d. Biology of the lymphatic system

128

e. Relation between the tumors and the lymphatic system

129

f. Metastasis of tumors

130

g. The sentinel node and distant lymph nodes

131

III. Staging Systems for Various Cancers

132

a. Colorectal cancer

132

b. TNM definitions for colorectal cancer

133

Stage 0

133

Stage I

134

Stage IIA

134

Stage IIB

134

Stage IIC

134

Stage IIIA

134

Stage IIIB

134

Stage IIIC

135

Stage IVA

135

Stage IVB

135

c. Breast cancer

135

d. Breast cancer in situ (DCIS and LCIS)

135

e. Invasive breast cancer

136

f. Definitions for breast cancer

136

g. Breast cancer staging

138

Stage 0

138

Stage IA

138

Stage IB

139

Stage IIA

139

Stage IIB

139

Stage IIIA

139

Stage IIIB

140

Stage IIIC

140

Stage IV

140

IV. Summary

141

V. The will Rogers Phenomenon

141

a. Will Rogers phenomenon for prostate cancer

141

b. Will Rogers phenomenon for non-small lung cancer

142

c. Will Rogers phenomenon for small cell lung cancer

142

d. Will Rogers phenomenon for rectal cancer

143

e. Will Rogers phenomenon for multiple sclerosis

143

VI. Other Sources of Artifacts in Data from Clinical Trials

144

VII. Concluding Remarks

144

6 Randomization, Allocation, and Blinding

146

I. Introduction

146

a. Allocation and allocation concealment

147

b. Simple randomization

149

c. Stratification

150

II. Manual Technique for Allocation

151

a. Allocation by coin-toss versus allocation by sealed envelope

153

III. Information on Randomization, Blinding, and Unblinding may be Included in the Clinical Study Protocol

154

a. Introduction

154

b. When to break the randomization code – clinical study protocol for trial on Alzheimer’s disease (27)

154

c. When to break the randomization code – clinical study protocol for trial on malaria vaccine (28)

155

d. When to break the randomization code – clinical study protocol for trial typhoid vaccine (29)

155

e. When to break the randomization code – clinical study protocol for trial on lung cancer (30)

155

f. When to break the randomization code – clinical study protocol for trial on sepsis (31)

156

g. When to break the randomization code – clinical study protocol for trial on melanoma (32)

156

h. When to break the randomization code – clinical study protocol for trial on multiple sclerosis (33)

156

IV. Summary

157

V. Subjects are Enrolled into Clinical Trials, One by One, Over the Course of Many Months

157

VI. Blocked Randomization

158

VII. Blinding

158

VIII. Interactive Voice Response Systems

160

IX. Concluding Remarks

164

7 Placebo Arm as Part of Clinical Study Design

166

I. Introduction

166

II. Hawthorne Effect

167

III. The No-Treatment Arm

167

IV. Physical Aspects of the Placebo

168

V. Active Placebo

168

VI. Subjects in the Placebo Arm May Receive Best Supportive Care Or Palliative Care

169

VII. Clash Between Best Supportive Care and the Endpoint of HRQoL

171

VIII. Ethics of Placebos

171

8 Intent to Treat Analysis vs. Per Protocol Analysis

178

I. Introduction

178

a. Definition of intent to treat analysis

178

b. Deviations and inconsistencies

179

II. ITT Analysis Contrasted with PP Analysis

181

a. ITT analysis vs. PP analysis – the Hosking study

182

b. ITT analysis vs. PP analysis – the Sethi study

183

c. ITT analysis vs. PP analysis – the Abrial study

183

d. ITT analysis vs. PP analysis – the Berthold study

183

e. ITT analysis vs. PP analysis – the Geddes study

184

f. ITT analysis vs. PP analysis – the Chauffert study

184

III. Disadvantages of ITT Analysis

184

IV. Run-in Period, as Part of the Study Design, is Relevant to ITT Analysis and PP Analysis

185

V. Summary

186

VI. Hypothetical Example Where Study Drug and Control Drug have Same Efficacy

186

VII. Modified ITT Analysis

187

a. Flow chart showing subjects included in the ITT analysis, modified ITT analysis, and PP analysis

188

b. Reasons for using modified ITT analysis

190

c. Excluding subjects who failed to meet inclusion or exclusion criteria, or who failed to receive study drug – the Vaira study

190

d. Excluding subjects who failed to meet inclusion or exclusion criteria – the Weigelt study

191

e. Excluding subjects who failed to meet inclusion or exclusion criteria – the Pinchichero study

191

f. Excluding subjects who failed to meet inclusion or exclusion criteria – the Leroy study

192

g. Exclusion of study subjects because of failure to satisfy the inclusion criteria, and for withdrawing consent – the Dupont study

193

h. Exclusion of study subjects because of failure to satisfy the inclusion criteria – the Florescu study

194

i. Excluding subjects who took prohibited drugs during the clinical trial, or who withdrew consent – the Manegold study

194

j. Excluding subjects who failed to receive the assigned treatment because of a mistake by the health care provider – the Berek study

194

k. Exclusion of study subjects who failed to take drug long enough to have the expected efficacy – the Krainick-Strobel study

195

l. Excluding subject who dropped out because of adverse events, and because of the bad flavor of the study drug – the Kreijkamp-Kaspers study

195

m. Excluding subjects who failed to receive the assigned treatment because of adverse events – the Caraceni study

196

n. Modified ITT group based on a subgroup of study subjects – the Gralla study

196

VIII. Start Date for Endpoints in Clinical Studies

197

IX. Summary and Conclusions

199

9 Biostatistics

200

I. Introduction

200

a. Kaplan-Meier plot

200

b. Examples of Kaplan-Meier plots – the Holm study

201

c. Censoring data

203

d. Hazard ratio

204

II. Definitions and Formulas

206

III. Data from the Study of Machin and Gardner

207

IV. Data Used for Constructing the Kaplan-Meier Plot are from Subjects Enrolling at Different Times

207

V. Sample Versus Population

209

VI. What can be Compared

210

VII. One-Tailed Test Versus Two-Tailed Test

211

VIII. P Value

212

IX. Calculating the P Value – a Working Example

215

X. Summary

221

XI. Theory Behind the Z Value and the Table of Areas in the Tail of the Standard Normal Distribution

221

XII. Statistical Analysis by Superiority Analysis Versus by Non-Inferiority Analysis

222

10 Introduction to Endpoints for Clinical Trials in Pharmacology

226

I. Introduction

226

a. Phase I clinical trial endpoints

226

b. Clinical endpoints

226

c. Surrogate endpoints

226

d. Relatively objective endpoints versus relatively subjective endpoints

228

e. Using multiple endpoints, and choosing the endpoint on which to base conclusions

229

f. In choosing endpoints keep in mind the eventual goals of the clinical trial

230

11 Endpoints In Clinical Trials on Solid Tumors – Objective Response

232

I. Introduction

232

a. Objective response using RECIST criteria

233

b. Objective response – Demetri’s example of partial response

238

c. Objective response – van Meerten’s example of partial response

240

d. Objective response – example of progressive disease

241

II. Studies Characterizing an Association Between Objective Response and Survival

242

III. Avoiding Confusion when Using Objective Response as an Endpoint

243

a. Date for beginning objective response measurements in two study arms, relative to start date of treatment

243

b. Where multiple measurements of objective response are taken, which measurement is used for analysis of efficacy?

244

c. How is it possible to obtain a meaningful value for objective response, or for endpoints (PFS, TTP) that comprise objective response?

245

d. Objective response is reported in terms of a “rate” and also as a “percent”

245

e. Drugs that are cytostatic and not cytotoxic may provide misleading results, where the endpoint of objective response is used

245

f. Use of different criteria (standards) for objective response, and the availability of updated criteria

246

12 Oncology Endpoints: Overall Survival and Progression-free Survival

248

I. Introduction

248

II. Comparing Contexts of Use and Advantages of Various Endpoints

249

a. Contrast between PFS and TTP

249

b. Excellence of PFS as an endpoint

250

c. Why progression-free survival may be preferred over overall survival

251

1. Confusion from effects of non-study drugs given to subjects who leave the trial

251

2. Collecting data on overall survival may require an extended follow-up period

251

3. Weakened conclusions, regarding efficacy of study drug, when the endpoint is keyed to a longer timeframe

252

4. Confusion from the multiplicity of causes of death

252

5. Ethical reasons

253

6. Need for premature halt of the trial, where the halt allows collection of data on progression-free survival, but prevents collection of data on overall survival

253

7. Conclusions arising from data on overall survival may be redundant with conclusions made from data on PFS

253

d. Why overall survival may be preferred over PFS

254

1. Overall survival is the gold standard

254

2. The date of the event that triggers PFS may be ambiguous, while the date that triggers overall survival is not ambiguous

254

e. Endpoint keyed to one specific time point – 6-month PFS

255

f. Use of the word “rate”

255

III. Data on Overall Survival and PFS from Clinical Trials

256

a. Utilities of the endpoints of objective response, PFS, and overall survival

256

1. Data on PFS may be more significant than data on overall survival – the Maemondo study

256

2. Methodology tip – shapes of Kaplan-Meier plots in the Maemondo study

259

3. Methodology tips – independent radiology assessments in the Gradishar study

259

4. The endpoint of PFS may have an advantage, where PFS data are more statistically significant than overall survival data – the Robert study

260

5. Rationale for combining trastuzumab with a platinum drug

262

6. Data on PFS can present earlier, and can be more dramatic, than data on overall survival – the Slamon study

263

7. Progression-free survival and subgroup analysis – the Van Cutsem study

266

8. Anti-sense drug for melanoma and subgroup analysis – the Bedikian study

269

IV. Summary

270

13 Oncology Endpoints: Time to Progression

272

I. Introduction

272

II. Agreement of Results from Objective Response, Time to Progression, and Overall Survival – the Paccagnella Study

273

III. Can the Value for PFS be Less than the Value for TTP?

273

IV. Time to Progression may be the Preferred Endpoint where, Once the Trial is Concluded, Patients Receive Additional Chemotherapy – the Park Study

274

V. The Endpoint of TTP may be Preferred Over Survival Endpoints, where Deaths Result from Causes Other than Cancer – the Llovet Study

275

VI. The Endpoint of Overall Survival may be Preferred Over Objective Response or Over TTP, where the Drug Is Classed as a Cytostatic Drug – the Llovet Study

277

VII. Time to Progression may Show Efficacy, where the Endpoint of Overall Survival Fails to Show Efficacy, where the Number of Subjects is Small – the McDermott Study

279

VIII. Time to Progression may Show Efficacy, where the Endpoint of Overall Survival Failed to Show Efficacy, where the duration of the Trial was too Short – the Cappuzzo Study

280

IX. Methodology TIP – Advantage of Using an Endpoint that Incorporates a “Median” Time

281

X. Summary

282

XI. Thymidine Phosphorylase as a biomarker for Survival – the Meropol Study

282

XII. Drug Combinations that Include Capecitabine

284

XIII. Methodology TIP – do Changes in mRNA Expression Result in Corresponding Changes in Expression of Polypeptide?

284

XIV. Conclusions

285

14 Oncology Endpoint: Disease-free Survival

286

I. Introduction

286

II. Difference Between Disease-Free Survival and Progression-Free Survival

287

III. Ambiguity in the Name of the Endpoint, “Disease-Free Survival”

288

IV. Disease-Free Survival Provides Earlier Results on Efficacy than Overall Survival – the Add-on Breast Cancer Study of Romond

289

V. Disease-Free Survival as an Endpoint in the Analysis of Subgroups – the Add-on Breast Cancer Study of Hayes

290

VI. Neoadjuvant Therapy Versus Adjuvant Therapy for Rectal Cancer – the Roh Study

292

VII. Where Efficacy of Two Different Treatments is the Same, Choice of Treatment Shifts to the Treatment that Improves Quality of Life – the Ring Study

293

VIII. Disease-Free Survival and Overall Survival are Useful Tools for Testing and Validating Prognostic Biomarkers – the Bepler Study

294

IX. Summary

295

15 Oncology Endpoint: Time to Distant Metastasis

298

I. Introduction

298

II. Time to Distant Metastasis Data are Acquired Before Overall Survival Data are Acquired – the Wee Study

299

III. Time to Distant Metastasis Data Can Reveal a Dramatic Advantage of the Study Drug, in a Situation Where Overall Survival Fails to Show Any Advantage – the Roach Study

301

IV. Use of a Gene Array as a Prognostic Factor for Breast Cancer Patients, Using the Endpoint of Time to Distant Metastasis – the Loi Study

302

V. Use of Micro-RNA Expression Data as a Prognostic Factor for Breast Cancer Patients – the Foekens Study

303

VI. Biology of Micro-RNA

304

VII. Conclusions

305

16 Neoadjuvant Therapy versus Adjuvant Therapy

306

I. Introduction

306

II. Advantages of Neoadjuvant Therapy

307

a. Killing micrometastases

308

b. Making surgery easier

308

c. Preserving functions, or cosmetic issues, of organs

308

d. Enables the physician to perform an experiment that enables a decision regarding subsequent therapy

309

e. Better ability of patient to tolerate chemotherapy

310

III. Advantages of Adjuvant Therapy

310

a. Immediate surgery and reduced risk of metastasis

310

b. More accurate staging

311

c. Drugs that require chronic treatment, for example for 5 years

311

IV. Two Meanings of the Term Adjuvant

311

V. Concluding Remarks

312

17 Hematological Cancers

314

I. Introduction

314

a. Classification of hematological cancers

314

b. Hematopoietic stem cells give rise to the lymphoid lineage and myeloid lineage

317

c. Locations of leukemic cells in the body

319

d. Lymphoid neoplasms

319

1. Acute lymphocytic leukemia

319

2. Chronic lymphocytic leukemia

322

3. Hairy cell leukemia

323

e. Myeloid neoplasms

324

1. Acute myeloid leukemia

324

2. Acute promyelocytic leukemia

326

3. Methodology tip – platelets and blood clotting

327

4. Chronic myeloid leukemia

328

II. Myelodysplastic Syndromes

329

a. Classifying MDS and scoring MDS

330

b. Treating MDS

331

c. Transfusions in MDS

332

d. Chromosome 5 abnormality and lenalidomide for treating MDS

333

e. Mechanism of action of lenalidomide

333

f. Mechanism of action of 5-aza-deoxycytidine

334

III. Summary

334

IV. Cytogenetics and the Hematological Cancers

334

a. Cytogenetics for diagnosis and prediction – AML

335

b. Cytogenetics for diagnosis and prediction – ALL

335

1. Numeric abnormalities in ALL

337

2. Structural abnormality t(9;22) (Philadelphia chromosome) in ALL

337

3. Structural abnormality t(1;19) in ALL

338

4. Structural abnormality t(12:21) in ALL

339

c. Cytogenetics for diagnosis and prediction – CML

339

d. Utility of the Philadelphia chromosome in diagnosis, drug target, and for assessing response

340

e. Cytogenetics for diagnosis and prediction – CLL

342

f. Cytogenetics for diagnosis and prediction – myelodysplastic syndromes

343

V. Chromosomal Abnormalities in Solid Tumors

345

VI. Clinical Endpoints and Examples from Clinical Trials

345

a. Endpoint of event-free survival

346

b. Endpoint of relapse-free interval

348

VII. Cytogenetics as a Prognostic Marker – The Grever Study of CLL

349

VIII. Minimal Residual Disease

351

a. Example of use of minimal residual disease and relapse – the scheuring study of philadelphia chromosome positive ALL

352

b. Example of use of minimal residual disease and event-free survival – the basso study of philadelphia chromosome negative ALL

353

c. Methodology tip – flow cytometry for assessing minimal residual disease

355

d. Using cells acquired after chemotherapy (not before chemotherapy) as a prognostic factor for long-term relapse – the cilloni study

355

e. Methodology tip – should biomarkers be measured before or after chemotherapy?

357

f. Example of use of minimal residual disease – the Grimwade study using PML-RAR-alpha fusion protein

357

IX. Confluence of Cytogenetics and Gene Expression

358

X. Conclusions

359

18 Biomarkers and Personalized Medicine

362

I. Introduction

362

a. Predictive markers versus prognostic markers

363

b. Including biomarker tests in the study design

365

c. Criteria for surrogate markers

366

d. Clinical trials focusing on utility of a biomarker

367

1. Biomarkers in breast cancer – the Stratton study

367

2. Biomarkers in breast cancer – the Vogel study

369

3. Methodology tip – fluorescent in situ hybridization (FISH) technique

371

4. Circulating tumor cells as a prognostic biomarker for colon cancer – the Cohen study

372

5. Methodology tip – circulating tumor cells as a biomarker

373

6. Cytokeratin as a soluble protein biomarker for colon cancer – the Koelink study

373

7. Tumor infiltrating T cells as a prognostic biomarker for colon cancer – the Galon study

374

8. Tumor infiltrating T cells as a prognostic biomarker for colon cancer – the Morris study

375

e. Lymphocytes can kill cancer cells, but lymphocytes can also cause cancer

375

II. Microarrays

376

a. Microarray used in ovarian cancer – the Spentzos study

377

b. Microarray used in colon cancer – the Wang study

378

c. Microarray used in liver cancer – the Hoshida study

379

III. C-Reactive Protein

380

a. Biology of C-reactive protein

380

b. C-reactive protein as a cancer biomarker

382

1. C-reactive protein and lung cancer – the Allin study

382

2. C-reactive protein and liver cancer – the Wong study

382

3. C-reactive protein and melanoma – the Findeisen study

383

c. Methodology tip – identifying new biomarkers by mass spectroscopy

384

d. C-reactive protein and atherosclerosis

384

IV. Concluding Remarks

387

19 Endpoints in Immune Diseases

390

I. Introduction

390

II. Multiple Sclerosis

390

a. Diagnosis

391

b. Endpoints

392

c. Timing for measuring endpoints

394

d. Primary endpoint

394

e. Multiple sclerosis functional composite (MSFC) score

395

f. Secondary endpoints

395

g. Introduction to MRI and detecting the onset of brain lesions

397

1. Example of MRI photograph

399

2. T2-weighted MRI

399

3. T1-weighted MRI

400

h. Results from the kappos study

401

III. Concluding Remarks

401

20 Endpoints in Clinical Trials on Infections

404

I. Introduction

404

II. Clinical and Immunological Features of Hepatitis C Virus Infections

404

III. Acute HCV Versus Chronic HCV

405

IV. Drugs Against Hepatitis C Virus

406

V. Immune Responses Against Hepatitis C Virus

408

VI. Kinetics of Hepatitis C Virus Infections

408

VII. Responders Versus Non-Responders

412

VIII. Endpoints in Clinical Trials Against Hepatitis C Virus

413

a. Endpoints of the McHutchison study

414

b. Endpoints of the Di Bisceglie study

414

IX. Biomarkers and Hepatitis C Virus

416

X. Concluding Remarks

417

21 Health-related Quality of Life

418

I. Introduction

418

II. Summary

420

III. HRQoL Instruments Take on Increased Importance, When Capturing Data on Adverse Events, or in Trials on Palliative Treatments

420

IV. Scheduling the Administration of HRQoL Instruments

421

V. HRQoL Instruments in Oncology

422

a. Introduction

422

b. Symptoms and functioning

423

c. Formats for disclosing HRQoL results

424

d. Colorectal cancer

425

e. Melanoma

428

f. Non-small cell lung cancer

430

1. The Shepherd study

430

2. The Bezjak study

431

3. The Bonomi study

431

4. Representative list of clinical trials

432

g. HRQoL in breast cancer

433

1. Where survival data are identical in both study arms, HRQoL data turn the tide – the Watanabe study

433

2. HRQoL data demonstrate that long-term treatment is well tolerated – the Muss clinical trial

433

VI. Decisions on Counseling; Decisions on Chemotherapy Versus Surgery

434

VII. Conclusions

434

22 Health-related Quality of Life Instruments for Immune Disorders

436

I. Introduction

436

II. Short Form SF-36 Questionnaire

436

a. Arthritis

439

b. Psoriasis

439

c. Crohn’s disease

439

d. Chronic obstructive pulmonary disease

440

e. Multiple sclerosis

440

III. HRQoL Instruments Specific for Multiple Sclerosis

440

a. The Rudick study

441

b. EDSS score versus HRQoL score

442

c. Interferon-alpha-2a – the Nortvedt study

442

d. Interferon-beta-1a – the Jongen study

443

e. Glatiramer acetate – the Zwibel study

443

f. Meditation training – the Grossman study

444

IV. Conclusions

444

23 Health-related Quality of Life Instruments and Infections

446

I. Introduction

446

II. Health-Related Quality of Life Instruments with Chronic Hepatitis C Virus

446

a. Example of hepatitis C virus HRQoL – the Mathew study

447

b. Concluding remarks

448

24 Drug Safety

450

I. Introduction

450

a. Overview of drug safety

451

b. Examples of adverse events

453

c. Anticipating adverse events in the design of clinical studies

454

d. Dose modification

455

II. Safety Definitions

458

a. Definitions from U.S. and European regulatory agencies

458

1. Adverse events

459

2. Serious adverse event

459

3. Adverse drug reaction (ADR)

460

4. Unexpected adverse drug reaction

460

5. Potential confusion in defining adverse events

460

b. Classification of adverse events as induced by disease versus induced by the study drug

461

c. Classification of adverse events by considerations used by statisticians

462

d. ITT analysis versus per protocol analysis

462

e. Summary

466

f. Classification of adverse events as anticipated versus unanticipated

466

g. Using raw data on adverse events to acquire cause-and-effect data on adverse drug reactions

470

III. Paradoxical Adverse Drug Reactions

471

a. Paradox with chemotherapy for cancer

472

b. Paradox with growth factors for cancer

473

c. Paradox with anti-depressants and depression

474

d. Paradoxes with drugs for treating bronchial constriction

475

IV. Monitoring and Evaluating Adverse Events

475

a. The data manager’s tasks include documenting missing data

476

b. CTCAE dictionary

477

c. Examples of missing data in documents submitted to the FDA

478

d. Writing style in case report forms

479

V. Adverse Events – Capturing, Transmitting, and Evaluating Data on Adverse Events

480

VI. Post-Marketing Report of Adverse Events

484

a. The MedWatch form, the yellow card, and the CIOMS I form

485

1. CIOMS

485

2. The CIOMS I form

486

b. Post-marketing surveillance

486

VII. Risk Minimization Tools

487

a. Introduction

487

b. Dear Healthcare Professional letter regarding birth control pills

490

c. Dear Healthcare Professional letter regarding acne medicine

491

d. Dear Healthcare Professional letter regarding appetite suppressants

492

VIII. Patient-Reported Outcomes

492

a. Introduction

492

b. PROs – example of head and neck cancer

493

IX. Summary of Reporting Systems Suitable for Capturing Adverse Events

495

X. Data and Safety Monitoring Committee

495

a. The DMC Charter

497

Data Safety Monitoring Board Charter

497

INTRODUCTION

498

ROLE OF THE BOARD

498

BOARD MEMBERSHIP

498

TERM

498

CONFLICT OF INTEREST AND FINANCIAL DISCLOSURE

499

COMPENSATION

499

BOARD MEETINGS AND REPORTS

499

ORGANIZATIONAL MEETING

499

INTERIM REVIEW MEETINGS

499

FORMAT

499

PARTICIPANTS

500

REVIEW MATERIALS

500

PERIODIC REPORTS TO THE DMC

501

UNSCHEDULED MEETINGS

501

DMC RECOMMENDATIONS

501

OUTSIDE EXPERTS

501

ACCESS TO INTERIM RESULTS

502

STOPPING RULES

502

COMMUNICATIONS

502

MEETING MINUTES

502

OTHER COMMUNICATIONS

502

SPONSOR’S DECISIONS AND ANNOUNCEMENTS

503

TIMETABLE

503

CONTACT INFORMATION

503

XI. Concluding Remarks

503

25 Mechanism of Action, Part I

506

I. Introduction

506

II. MOA and the Package Insert

507

III. MOA and Surrogate Endpoints

508

IV. MOA and Expected Adverse Drug Reactions

508

V. MOA and Drug Combinations

509

a. Drug combinations that are complementary or synergic

509

b. Drug combinations that avoid inducing cross-resistance

509

VI. Mechanism of Action of Diseases with an Immune Component

510

a. Introduction

510

b. Diseases with an immune component

511

c. Messengers in the immune system

511

d. Cells of the immune system

513

e. Processing and presentation of antigens, T cell activation, and T cell maturation

517

f. Drugs that modulate the immune system

517

1. Vaccines

518

2. Cytokines

518

3. TLR-agonists

519

4. Methodology tip – fine tuning of immune adjuvants when treating cancer

520

5. Antibodies

521

6. Treg inhibitors

521

VII. Immunology can be Organized as Pairs of Concepts

522

a. Myeloid DCs and plasmacytoid DCs

523

b. Th1-type response and Th2-type response

523

c. Externally acquired antigens and internally acquired antigens

523

d. Polypeptide antigens can contain both MHC class I and MHC class II epitopes

524

e. CD4+ T cells and CD8+ T cells

524

f. Two different mechanisms of CTL response

524

g. Naive response and memory response

524

h. Specific immunity and innate immunity

525

VIII. Conclusions

525

26 Mechanisms of Action, Part II – Cancer

528

I. Immune Response Against Cancer

528

a. Mechanisms of immune response against tumors

529

b. Natural killer cells and antibody-dependent cell cytotoxity

532

c. Regulatory T cells

534

d. Concluding remarks

536

27 Mechanisms of Action, Part III – Immune Disorders

538

I. Introduction

538

a. Mechanisms of action summaries for various immune disorders

539

1. Rheumatoid arthritis

540

2. Psoriasis

540

3. Lupus

540

4. Crohn’s disease and ulcerative colitis

541

5. Asthma

541

6. Chronic obstructive pulmonary disease (COPD)

541

II. Detailed Example of Multiple Sclerosis Mechanism of Action

541

a. Natalizumab

542

b. Fingolimod

542

c. Interferon-beta1 (IFNbeta1)

543

d. Cladribine

544

e. Animal model for multiple sclerosis

545

f. Mechanisms leading to multiple sclerosis are complex and not firmly established

546

g. Lesions of multiple sclerosis in humans

546

1. Initiating events in multiple sclerosis

546

2. CD8+ T cells attack nerves

546

3. Contributions of CD4+ T cells

547

4. Dendritic cells present antigen to T cells and activate the T cells

547

5. Breakdown of blood–brain barrier

548

6. Toxic oxygen from microglia

548

7. Diagram of multiple sclerosis

548

III. Concluding Remarks

550

28 Mechanisms of Action, Part IV – Infections

552

I. Introduction

552

II. Hepatitis C Virus Infections

552

a. Protease inhibitors used as drugs against anti-hepatitis C virus

553

b. Mechanisms of immune response against hepatitis C virus antigens

555

c. Methodology tip – GenBank

557

d. Dendritic cells and antigens of hepatitis C virus

558

e. Hepatitis C, chronic inflammation, and liver cancer

560

f. Dendritic cells

560

g. Sources of interferons during HCV infections

560

h. What IFN-gamma does during HCV infections

561

i. What T cells do during HCV infections where the patient spontaneously recovers

561

j. What immune cells do during HCV infections where the patient develops a chronic HCV infection

562

k. In HCV infections, IL-12 stImulates NK cells to express IFN-gamma

562

l. In HCV infections, IFN-alpha stimulates NK cells (or CD8+ T cells) to express IFN-gamma

563

m. Influence of IFN-alpha on gene expression as measured by microarrays

565

n. Diagrams of the immune network in immune response against HCV

566

o. Methodology tip – populations of leukocytes in the bloodstream

566

III. Concluding Remarks

568

29 Consent Forms

570

I. Introduction

570

a. An early clinical study using a consent form – yellow fever study

570

b. The consent form of the Yellow Fever Commission

572

c. Summary

573

II. Sources of the Law in the United States

573

III. Guidance for Industry

574

IV. Ethical Doctrines

575

V. The Case Law

576

VI. Basis for Consent Forms in the Code of Federal Regulations

576

VII. Summary

578

VIII. Examples of Contemporary Consent Forms

578

a. Example of a contemporary consent form (reproduced in full) (39,40)

579

b. Another example of a contemporary consent form (reproduced in part)

584

c. Comparison of standard consent form with the more elementary consent form

586

d. Analysis of consent forms by the medical community

587

e. Most consent forms are written at a level that is too advanced

588

IX. Ethical Issues Specific to Phase I Clinical Trials in Oncology

590

X. Decision Aids

591

XI. Distinction Between Stopping Treatment and Withdrawing from the Study

593

XII. Concluding Remarks

593

30 Package Inserts

596

I. Introduction

596

a. FDA’s Guidance for Industry documents relating to package inserts

597

b. Classes of drugs

600

c. Black box warning

600

d. Summary

602

II. Potential Ambiguity of Writing in Package Inserts

602

III. Package Insert may Protect Manufacturer from Liability

603

a. Opinion concerning dicumarol

604

b. Opinion regarding kanamycin

605

c. Opinion regarding dilantin

605

d. Opinion concerning oxytocin

605

e. Opinion regarding oral polio vaccine

606

f. Opinion regarding norethindrone

607

g. Summary

607

IV. Package Insert Compared with Consent Form

608

V. Relation between Package Inserts to the Standard of Care, and to off-Label Uses

608

VI. Conclusions

610

31 Regulatory Approval

612

I. Introduction

612

a. Origins of the Federal Food, Drug and Cosmetic Act and its amendments

612

b. Federal Food, Drug and Cosmetic Act of 1938

613

c. Drug Amendments Act of 1962

615

d. Food and Drug Administration Modernization Act of 1997 and Phase IV clinical trials

615

II. History of the European Medicines Agency

616

III. International Conference on Harmonisation

618

IV. History of the Medicines and Healthcare Products Regulatory Agency

620

V. Outline of Regulatory Approval in the United States

621

a. The Investigational New Drug

621

b. The Investigational New Drug and the Common Technical Document

626

VI. Process of Administering Clinical Trials

627

VII. Process of Medical Writing

630

a. Grammatical issues

631

b. Formatting issues

632

VIII. Meetings with the U.S. Food and Drug Administration

635

a. Introduction

635

b. Paper trail of FDA’s decision-making process for individual drugs

636

c. Clinical review

636

d. Pharmacology review

638

e. Approval letter

638

f. Snapshots of the FDA’s regulatory review process

639

1. Example showing transition from an open-label Phase I trial to a blinded Phase II trial

639

2. Example showing how FDA uses data from Phase I trial to arrive at a dose for using in a Phase II trial

640

32 Patents

642

I. Introduction

642

a. History of patenting

642

b. Outline of the patenting process

644

c. Summary

645

II. Types of Patent Documents

646

III. Structure of Patents

647

a. Introduction

647

b. The claims

648

IV. Timeline for Patenting

651

V. Sources of the Law for Patenting

654

VI. Intersections between the FDA Review Process and Patents

656

a. Introduction

656

b. Using patent as source documents when writing regulatory submissions

657

Index

660