active pharmaceutical ingredients

354

Upload: ivana-veselinovic

Post on 11-Nov-2014

242 views

Category:

Documents


38 download

DESCRIPTION

Active pharmaceutical ingredients known today as ‘‘APIs’’ areorganic chemicals, generally synthetic, that are the subject ofthis book. These ingredients are chemicals that will be usedin a final pharamaceutical dosage form...

TRANSCRIPT

Page 1: Active Pharmaceutical Ingredients
Page 2: Active Pharmaceutical Ingredients

Active PharmaceuticalIngredients

Development, Manufacturing, and Regulation

edited by

Stanley H. NusimS. H. Nusim Associates, Inc.

Aventura, Florida, U.S.A.

Boca Raton London New York Singapore

DK2287_half-series-title.qxd 4/14/05 12:35 PM Page i

Page 3: Active Pharmaceutical Ingredients

Published in 2005 byTaylor & Francis Group 6000 Broken Sound Parkway NW, Suite 300Boca Raton, FL 33487-2742

© 2005 by Taylor & Francis Group, LLC

No claim to original U.S. Government worksPrinted in the United States of America on acid-free paper10 9 8 7 6 5 4 3 2 1

International Standard Book Number-10: 0-8247-0293-X (Hardcover) International Standard Book Number-13: 978-0-8247-0293-9 (Hardcover)

This book contains information obtained from authentic and highly regarded sources. Reprinted material isquoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable effortshave been made to publish reliable data and information, but the author and the publisher cannot assumeresponsibility for the validity of all materials or for the consequences of their use.

No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, andrecording, or in any information storage or retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, please access www.copyright.com(http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood Drive,Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registrationfor a variety of users. For organizations that have been granted a photocopy license by the CCC, a separatesystem of payment has been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used onlyfor identification and explanation without intent to infringe.

Library of Congress Cataloging-in-Publication Data

Catalog record is available from the Library of Congress

Visit the Taylor & Francis Web site at http://www.taylorandfrancis.com

Taylor & Francis Group is the Academic Division of T&F Informa plc.

DK2287_Discl.fm Page 1 Tuesday, April 12, 2005 2:45 PM

Page 4: Active Pharmaceutical Ingredients

Preface

Active pharmaceutical ingredients known today as ‘‘APIs’’ areorganic chemicals, generally synthetic, that are the subject ofthis book. These ingredients are chemicals that will be usedin a final pharamaceutical dosage form. The manufacturingof these chemicals is a subsection of fine chemical manufactur-ing. This subsection of the chemical industry has undergonevery significant changes in much the same manner, butperhaps trailing, the pharmaceutical industry that manufac-tured the final dosage form.

The ‘‘pharmaceutical industry’’ at the turn of the 20thcentury was essentially the local pharmacy (or ‘‘chemist’’ asit was also known). The ‘‘bulk pharmaceutical chemical indus-try’’ at that time was merely a provider of all those laboratorychemicals, including solvents and excipients as well as APIsneeded by the local pharmacist to compound the prescribingdoctor’s formulation.

Over this past century, as with many industries, enor-mous changes have occurred in the pharmaceutical industry,causing equally significant changes for API suppliers. It isthese changes, many of which have accelerated in recentdecades, that suggested the need for a definitive referencefor this manufacturing activity.

iii

Page 5: Active Pharmaceutical Ingredients

At one time following routine chemical manufacturingpractices would have been sufficient; however, this is nolonger the case. Not only has there been a significant shiftin the government regulations that control the redefined‘‘quality’’ of the product, but a very intensive look at the devel-opment of the process to be used as well as the manufacturingactivities required to make the API.

This focus is to ensure that the API is produced in anenvironment that ensures it is free of contamination thatmay be introduced from inherent process impurities but alsofrom the manufacturing environment itself. The latter is con-trolled by the so-called ‘‘cGMPs’’ (current Good ManufacturingPractices), while the former by the nature of the chemical pro-cess and the level of quality assurance that the process pro-vides; hence, a focus on the process development is essential.

It is the intent of this volume to focus on the three overallactivities that bring an API to market; the development of thechemical process, the manufacturing activity utilizing thatprocess, and the governmental regulations that control theapproval of the product so that it may be commercially mar-keted. This book brings together information into a singlesource that will allow those in the field to be sure they areup to date. In addition, it will provide to those organizationsthat are planning to enter this field, the basic informationneeded to think through, understand, and effectively planbulk manufacturing of an API.

The rapidly changing environment that has occurred inthe past decades shows no signs of easing; thus, this volumewill be a starting point. Ongoing continuing attention to allaspects of these issues is an absolute necessity to ensure thatmanufactured APIs will meet the newest standards in anenvironment that has seen many changes in the market itselfas well as its regulation, product mix, and volume.

This text covers those three activities of development,manufacturing, and regulation in its broadest sense. This willinclude discussions on the process development cycle,introduction of the process into factory design engineering,regulatory matters that include the regulatory approval pro-

iv Preface

Page 6: Active Pharmaceutical Ingredients

cess, quality control=assurance, and validation as well as thestandard plant manufacturing operation activities includingmaterials management and planning and maintenance. Inaddition, it will discuss other plant operational issues includ-ing safety and environmental issues that are part of any che-mical manufacturing operation.

I have chosen to exclude fermentation and other biologi-cal processes from this book although products from thoseprocesses continue to be an increasingly important source ofpharmaceutical actives in today’s world. This decision wasmade because the chemical routes remain the largest sourceof actives to the pharmaceutical industry. Actives suppliedby biological processes are no less important than chemicallygenerated actives but are sufficiently different to be worthy oftheir own volume.

I wish to express my thanks to the publisher for its invi-tation to assemble this book and particularly to SandraBeberman for bearing with me in the very long and tediousdevelopment process for the book. Her advice and encourage-ment throughout this process was a primary driving force toensure its completion.

Stanley H. Nusim

Preface v

Page 7: Active Pharmaceutical Ingredients
Page 8: Active Pharmaceutical Ingredients

Contents

Preface . . . . iii

Contributors . . . . xiii

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1Stanley H. Nusim

I. Consolidation and Integration . . . . 2II. Quality . . . . 2

III. Potency . . . . 5IV. Computer Control and Automation . . . . 6V. Summary . . . . 7

2. Process Development . . . . . . . . . . . . . . . . . . . . . 9Carlos B. Rosas

I. Introduction . . . . 9II. The Bulk Drug Process as Part of the

Drug Development Program . . . . 13III. From the Bench to the Pilot

Plant and Beyond . . . . 39IV. The Physicochemical Attributes of the

Bulk Drug . . . . 61V. The Process Body of Knowledge . . . . 65

VI. Processing Responsibility in Bulk DrugProcess Development . . . . 73

VII. Outsourcing in Bulk DrugProcess Development . . . . 89

vii

Page 9: Active Pharmaceutical Ingredients

VIII. In Closing . . . . 89References . . . . 90

3. Bulk Drugs: Process Design, Technology Transfer,and First Manufacture . . . . . . . . . . . . . . . . . . . 93Carlos B. Rosas

I. Introduction . . . . 93II. The Process Design Task in Bulk

Drugs . . . . 96III. Technology Transfer of the Bulk Drug

Process and First Manufacture . . . . 106IV. In Closing—The Processing Technologies of

Bulk Drugs . . . . 123References . . . . 125

4. Design and Construction of Facilities . . . . . . . 127Steven Mongiardo and Eugene Bobrow

I. Introduction . . . . 127II. Business Requirements . . . . 129

III. Developing the Preliminary Scope . . . . 132IV. Utilities and Building Systems . . . . 137V. Preliminary Scope Deliverables . . . . 138

VI. Design Development . . . . 142VII. Utilities . . . . 147

VIII. Safety . . . . 150IX. Current Good Manufacturing Practices

Requirements . . . . 150X. Qualification Plan . . . . 151

XI. Expansion Capabilities . . . . 152XII. Hazard and Operability Analysis . . . . 152

XIII. Execution Strategy and Planning . . . . 154XIV. Procurement Strategy . . . . 156XV. Construction Management . . . . 161

XVI. Start–Up Acceptance . . . . 162XVII. Project Turnover and Installation

Qualification . . . . 163XVIII. Conclusions . . . . 165

References . . . . 166

viii Contents

Page 10: Active Pharmaceutical Ingredients

5. Regulatory Affairs . . . . . . . . . . . . . . . . . . . . . . . 167John Curran

I. Introduction . . . . 167II. Requirements for Submission of

Regulatory CMC Documents . . . . 169III. Contents of Regulatory

Submissions—API Sections . . . . 173IV. Registration Samples . . . . 191V. The Review and Approval Process . . . . 192

VI. Preapproval Inspections . . . . 195VII. Postapproval Change Evaluations . . . . 196

VIII. The Future . . . . 199IX. Helpful References . . . . 200

6. Validation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203James Agalloco and Phil Desantis

I. History . . . . 203II. Definition of Validation . . . . 205

III. Regulations . . . . 206IV. Application of Validation . . . . 206V. Life Cycle Model . . . . 206

VI. Validation of New Products . . . . 207VII. Validation of Existing Products . . . . 208

VIII. Implementation . . . . 209IX. Bulk Pharmaceutical Chemical

Validation . . . . 211X. In-Process Controls . . . . 220

XI. Cleaning Validation . . . . 222XII. Computerized Systems . . . . 225

XIII. Procedures and Personnel . . . . 225XIV. Validation of Sterile Bulk Production . . . . 225XV. Conclusion . . . . 231

References . . . . 231

7. Quality Assurance and Control . . . . . . . . . . . . 235Michael C. VanderZwan

I. Introduction . . . . 235II. Defining and Assuring the Quality of the Active

Pharmaceutical Ingredient . . . . 240

Contents ix

Page 11: Active Pharmaceutical Ingredients

III. The Regulations for Quality . . . . 245IV. The Quality Control and Quality

Assurance Department . . . . 273Appendix A . . . . 280

8. Plant Operations . . . . . . . . . . . . . . . . . . . . . . . . 285Stanley H. Nusim

I. Plant Organization . . . . 285II. Batch vs. Continuous . . . . 286

III. Dedicated vs. Shared ManufacturingFacilities . . . . 288

IV. Shift Operations . . . . 288V. Sterile Operations . . . . 291

VI. Clean Room . . . . 294VII. Cost Control . . . . 296

VIII. Fixed Overhead Absorption . . . . 299IX. Safety . . . . 300X. Environmental . . . . 303

9. Materials Management . . . . . . . . . . . . . . . . . . . 305Victor Catalano

I. Introduction . . . . 305II. Production Planning . . . . 306

III. Inventory Management . . . . 307IV. Purchasing=Supply Management . . . . 308V. Distribution=Transportation . . . . 315

VI. Information Technology . . . . 316VII. Quality Management . . . . 317

References . . . . 318

10. Plant Maintenance . . . . . . . . . . . . . . . . . . . . . . 323Raymond J. Oliverson

I. Introduction . . . . 323II. Strategic Plan . . . . 324

III. Reliability-Balanced Scorecards . . . . 325IV. Maintenance Basics . . . . 327V. Condition Monitoring . . . . 329

VI. Operator-Driven Reliability . . . . 330

x Contents

Page 12: Active Pharmaceutical Ingredients

VII. Reliability Engineering . . . . 331VIII. Risk Management . . . . 333

IX. Summary . . . . 334

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 335

Contents xi

Page 13: Active Pharmaceutical Ingredients
Page 14: Active Pharmaceutical Ingredients

Contributors

James Agalloco Agalloco & Associates, Inc., Belle Meade,New Jersey, U.S.A.

Eugene Bobrow Merck & Co., Inc., Whitehouse Station,New Jersey, U.S.A.

Victor Catalano Purchasing Group Inc. (PGI), Nutley,New Jersey, U.S.A.

John Curran Merck & Co., Inc., Whitehouse Station,New Jersey, U.S.A.

Phil Desantis Schering-Plough Corp., Kenilworth,New Jersey, U.S.A.

Steven Mongiardo Merck & Co., Inc., Whitehouse Station,New Jersey, U.S.A.

Stanley H. Nusim S. H. Nusim Associates Inc., Aventura,Florida, U.S.A.

Raymond J. Oliverson HSB Reliability Technologies,Kingwood, Texas, U.S.A.

xiii

Page 15: Active Pharmaceutical Ingredients

Carlos B. Rosas Rutgers University, New Brunswick,New Jersey, U.S.A.

Michael C. VanderZwan Pharmaceutical Technical,Roche Pharmaceuticals, Basel, Switzerland

xiv Contributors

Page 16: Active Pharmaceutical Ingredients

1

IntroductionSTANLEY H. NUSIM

S. H. Nusim Associates Inc., Aventura, Florida, U.S.A.

I. Consolidation and Integration . . . . . . . . . . . . . . . . . . 2II. Quality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2III. Potency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5IV. Computer Control and Automation . . . . . . . . . . . . . . . 6V. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7

Pharmachemical manufacturing is that branch of fine chemi-cal manufacturing that is directed to the manufacture ofchemicals whose ultimate use will be in a final pharmaceuticaldosage form, referred to as the active pharmaceutical ingredi-ent (‘‘API’’). This industry segment has undergone very signif-icant changes in much the same manner, but trailing, thepharmaceutical industry itself, from the time it emerged earlyin the 20th century.

Thus, we must examine what has happened in the phar-maceutical industry over this period, in order to understandthe implications for API manufacturing. This will lead us tothe present time and to the goal of this book.

It is our objective to provide a reference book that speaksto those issues that need to be addressed in order to assure thatan existing or proposed pharmachemical operation will meetits objective of supplying an API to meet a medical=marketneed efficiently and effectively.

1

Page 17: Active Pharmaceutical Ingredients

The changes are themselves a result of major changesthat have occurred both directly and indirectly, in and on,the industry. These changes include company consolidations,both backward and forward integration, the increased andchanged role of quality, the significant intensification of regu-latory bodies worldwide, the impact of the greatly increasedpotency of APIs, thereby reducing pharmachemical require-ments and the broadening of the market worldwide.

These ideas will be discussed briefly here and touched onin depth in the subsequent chapters.

I. CONSOLIDATION AND INTEGRATION

The ‘‘pharmaceutical industry’’ at the turn of the 20th centurywas essentially the local pharmacy (or chemist as it was alsoknown outside of the United States). The objective of the phar-machemical supplier to the local industry, at that time, was aprovider of all the chemicals, including APIs, as needed by thepharmacist to compound the prescribing doctor’s prescription.

Thus, the great pharmaceutical titans of today, such asMerck,were afine chemicalmanufacturer providing a full vari-ety of basic laboratory chemicals and solvents as well as theactives of the day, in order to meet all of the formulating needsof the pharmacist. This activity was common in those earlydays, as well, to Pfizer, Bayer, and Sterling, among others.

The forward integration of these companies into provid-ing the finished dosage form had by the middle of this pastcentury become the standard rather than the exception, asthe medical community shifted to writing prescriptions forthe local pharmacist to fill prescribing finished dosage formsrather than their own formulations. This practice continuestoday, as the determination of efficacy and safety of formu-lated product has grown to very significant proportions.

II. QUALITY

An overriding driving force in this direction, although it maynever have been originally intended, has been the shift of

2 Nusim

Page 18: Active Pharmaceutical Ingredients

governmental control that has been exercised by the U.S.Food & Drug Administration (‘‘FDA’’). A brief discussion ofthat change is now in order.

The initial purpose of the first Pure Food, Drug &Cosmetics Act (Act) that was passed by Congress in the firstdecade of the last century was one of safety. It began by theregulation of those items of commerce that had the potentialof poisoning the individual who used it if the product wascontaminated. It is for this reason that the Act coveredthose three specific items, all lumped together although eachbeing used for very different purposes.

The initial focus, at that time, for drugs as well as theother two types of ingested or topically applied products,was lack of contamination as determined by quality samplingand testing. In addition, and extrapolating that issue to newproposed pharmaceuticals, the key data required was thetoxicity data and its ratio to the proposed dose level, the ‘‘ther-apeutic index.’’ However, no data or judgment on efficacy wasrequired for its proposed use. Its medical purpose and itsultimate use remained in the hands of the physician andthe sponsoring company that promoted it.

In the middle 1950s, this changed dramatically when theAct was amended significantly. The change, driven bycongressional hearings and the ‘‘Thalidomide affair,’’a

now required not only more significant safety data, beyondsimple toxicity, but also, more significantly, scientific proof

aThalidomide was an antinausea drug that at that time hadbeen approved in Europe and was before the FDA forapproval in the United States. Pregnant women who are nor-mally prone to nausea became an instant market for the newdrug. However, very serious birth defects (missing limbs)were experienced in babies born to many of those womenwho had taken the drug. This precipitated a worldwide reac-tion to review the new drug approval process. Needless to saythat the drug was not approved in the United States at thattime. (In recent years, it has been approved for limited specialuse in leprosy.)

Introduction 3

Page 19: Active Pharmaceutical Ingredients

of efficacy. This now placed a new burden on the sponsoringcompany to provide unequivocal proof, to the government’ssatisfaction, that the addition of a new chemical entity atthe dose level recommended was worthwhile to the public.The shift was due to the recognition that replacing a triedand true medication, that was widely used and its side effectswell defined with a new compound with only limited experi-ence in man, was in itself an unknown risk and, therefore,must be shown to be worth the risk.

This greatly increased the cost and the risk associatedwith the discovery and introduction of new chemical enti-ties. This change was absorbed by the industry and setthe stage for the next major shift in policy that came inthe middle 1970s. This was the establishment of currentgood manufacturing practices (cGMPs) for the manufactureof pharmaceutical actives as well as the finished pharmaceu-tical products.

This was the next step in the focus of the FDA on thesafety of the product. Up until this point, contamination (orlack thereof) was defined by the presence (or absence) offoreign impurities not specified in the analytical protocol forthe product. This was the case for either the pharmaceuticalproduct or the API that went into the finished product.Although this could be a definitive test for a uniformly distrib-uted contaminant, it would not necessarily find randomcontamination that occurred in processing or extraneousmatter that could enter the system from dirty facilities or pooroperating practices.

Finished goods testing, today, as it was at that time alwaysdepended upon the assumption of uniformity of product. It wasthis presumption that permitted the approval and release of aproduct based on the testing of 100g of a 100kg pharmachem-ical batch or 30 tablets of a lot of 500,000 tablets.

The concept of current ‘‘cGMPs’’ and quality assurancebecame the dominant theme, thereby pushing the analyticaltesting (quality control) into the background.

In principle, one now had to show, in order to have a pro-duct free of contamination, that the manufacturer producedthe product in contaminant-free equipment in a clean facility,

4 Nusim

Page 20: Active Pharmaceutical Ingredients

within equipment designed and tested to show consistent andreproducible product by people thoroughly trained and withfull knowledge of the process. Thus, in the United States, thisgreatly shifted the emphasis to a more rigorous standard of‘‘quality’’.

The most recent change implemented is the requirementof formal ‘‘validation’’ of facilities, equipment and the processitself. This is the ‘‘proof’’ that the process and the facility canproduce quality product on a consistent basis.

In a similar fashion, one can see the extension of thetighter regulations as they apply in the United States, toJapan and Western Europe. Through the EU, they haveimplemented similar standards for the very same reason inEurope; additionally, many of the ‘‘third world’’ nations havealready implemented their own GMP initiatives reemphasiz-ing the growing uniformity in such requirements throughoutthe world.

All these factors are discussed more thoroughly in theappropriate chapters within this book.

III. POTENCY

A subtle change that has emerged in the methods of discover-ing and developing new drugs in the past decades has hadsignificant impact on the pharmachemical industry.

In the early days, the key to drug discovery often wasscreening programs, where laboratory-screening models wereused to test new chemical entities for efficacy against specificdisease candidates. Those that were effective, however, oftenfound much of their potency diminished as the active, gener-ally formulated into a pill, was attacked by normal bodychemistry as it passed through the digestive system on itsway to be absorbed into the blood and transported to the dis-ease site. Thus, only a fraction of the orally ingested drugreached the drug target area. As a result, dose regimens formost oral drugs were 100–500mg.

These dosing levels generated needs for significant quan-tities of actives in some cases into the millions of kilograms

Introduction 5

Page 21: Active Pharmaceutical Ingredients

annually. (Five billion tablets at 200mg dose require 1 millionkilograms of active.) This resulted in dedicated plants for eachdrug active, particularly since the active was generally a com-plex organic molecule requiring many chemical steps tosynthesize.

However, with the advent of the focus on biochemistryand the new sophistication gained in understanding thechemistry and biology of the body, today’s drugs are designedso as to be more potent. In addition, they can be chemicallyprotected to limit the destruction of the drug as it passesthrough the body on its way to the target site. Thus, normaldosing of today’s ‘‘designer’’ drugs are 5–20mg, 10 fold lessthan in the past. This reduces the API need for ‘‘blockbuster’’drugs by an order of magnitude (5 billion tablets at 10mg doserequires 50,000kg of API). This also suggests that those les-ser volume products would require very small quantities ofAPI making dedicated facilities for them very uneconomical.

These factors have refocused API manufacturing fromfacilities dedicated to a single API product to multiproductmanufacturing facilities. The added costs of a facility due tothe more rigorous cGMPs that now apply favor these kindsof facilities, where the cost can be shared by many rather thana single product.

This adds a very critical aspect to the operation becausethe issues of equipment clean out and turnaround, particu-larly as the issue of cleanliness to ensure that cross contami-nation does not occur must be addressed.

IV. COMPUTER CONTROL ANDAUTOMATION

This industry, like nearly all others, has seen the positiveimpact of the introduction of computers and automation inthe manufacturing facilities. The first impact was in the auto-matic control systems that are used to maintain accurate andreproducible operating conditions for reaction and isolationsystems. This was extended into the integration of multipleoperations under computer control often eliminating or atleast minimizing people intervention.

6 Nusim

Page 22: Active Pharmaceutical Ingredients

This itself caused some concerns for the FDA, which, inthe past, depended upon manual documentation by operatorsof batch procedures written and issued by people and peopleobserving and recording all data. This was transformed tocomputer-recorded data and operating instructions beingmaintained in computer files. This generated an entire seriesof new issues that had to be dealt with by both the operationand the FDA. First was security to be sure that the automatedinstructions are safe from improper and unauthorizedchanges to the issue of signatures, often electronic signatures,a new concept that has become very common.

V. SUMMARY

The changes referred to above, and the changes that are tooccur, without doubt, in the future, drive the need to under-stand where we are today and where we are going in thefuture. We have chosen to address the various segmentsand activities of a pharmachemical plant by having a focuseddiscussion on each in the subsequent chapters.

Again, I repeat a statement from the Preface. Each andevery topic covered in this volume has changed from the pastand will continue to change in the future; therefore, thereader is being presented with a ‘‘starting point’’ from whichhe or she must continue to follow the progress of in order tokeep current.

Introduction 7

Page 23: Active Pharmaceutical Ingredients
Page 24: Active Pharmaceutical Ingredients

2

Process DevelopmentCARLOS B. ROSAS

Rutgers University, New Brunswick, New Jersey, U.S.A.

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9II. The Bulk Drug Process as Part of the Drug Development

Program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13III. From the Bench to the Pilot Plant and Beyond . . . . . . 39IV. The Physicochemical Attributes of the Bulk Drug . . . . . 61V. The Process Body of Knowledge . . . . . . . . . . . . . . . 65VI. Processing Responsibility in Bulk Drug Process Development73VII. Outsourcing in Bulk Drug Process Development . . . . . 89VIII. In Closing . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90

I. INTRODUCTION

The purposes of this chapter are few and rather ambitious.First, to provide a sound perspective of bulk drug process workto the uninitiated and the relatively new practitioner, hope-fully without prejudice to the benefit that the approach hereinmight afford to an experienced but still restless practitioner.All work in a forest that is dense and rich in its variety; itshould be regarded from a vantage now and then, and it isfrom such a deliberately selected vantage that the chapterunfolds.

Then there is the promotion of the power that the purpo-seful convergence of chemistry, microbiology, and chemical=

9

Page 25: Active Pharmaceutical Ingredients

biochemical engineering can bring to bear on the increasinglydifficult task at hand: the timely conception, development, andreduction to practice at scale of a sound process for the manu-facture of a bulk drug. In the 2000s, timely is shorthand forswift, sound encompasses safety to the environment and topeople as well as amenable to various regulatory approvals,and reduction to practice at scale means that the resultingprocess can be used for reliable manufacture in whatevercontext might be first required.

Chemistry, in the context at hand, is the aggregate ofsynthetic, analytical, and physical chemistry fieldswithin what may be called the drug process chemistrydiscipline at large. The latter, while practiced for dec-ades, has truly come into being in the 1990s, spurredmostly by the greater ascendance of the pharmaceuticalindustry among chemistry practitioners and by theenhanced role of the bulk drug process in the outcomeof drug development. Whereas toxicology or clinicalresults were the exclusive causes for the demise of drugcandidates, the greater difficulty in making today’s morecomplex structures in today’s regulatory milieu has forsome time raised the profile of their bulk process develop-ment task as a factor in the overall outcome (1).

Although first manufacture of the bulk drug is the para-mount objective of the technology transfer to manufac-turing, the process body of knowledge should be sturdyand complete enough to support expanded manufacturefor product growth, as well as provide at least a clearsense of direction for process improvements or second-generation processing.

The above definitions conveniently describe a complextask, to which considerable skills need to be applied withdue deliberation and under constant managerial attention.Indeed, successful bulk drug process development, as justdefined, requires that sufficient interdisciplinary and opera-tional resources be brought together in a cohesive manner,not unlike that required by a critical mass in nuclear fission.Most often, having the resources is not enough, and theircohesiveness makes a significant difference in the degree of

10 Rosas

Page 26: Active Pharmaceutical Ingredients

success, sometimes making the ultimate difference: having ornot having a new drug available when needed.

Another sought perspective applies to the integration ofthe bulk drug process development task with the simultaneousdrug development program at large: toxicology, dosage formdevelopment, clinical development, and the assembly of theregulatory submissions. The latter, leading to the desiredregulatory approvals as the culmination of the overall effort,has in recent years become increasingly dependent on the scopeand execution of the process work for the bulk drug, which insome of its aspects has now become fastidious and greatlyincreased the burdens of the bulk process development task.

As the last objective, the methods of bulk drug processdevelopment will be weaved discreetly, if not seamlessly,throughout the chapter: (a) the principal issues that shapethe methods, (b) the most trenchant choices confronting theprocess development team, and (c) some selected heuristics(i.e., empirical rules that, although lacking proof, are usefuloften enough) distilled from the author’s experience.

As a distinct and credible literature of process develop-ment for bulk drugs and fine chemicals has come intobeing and grows, statements of applicable empirical wis-dom are appearing with a modicum of organization (2–6)and the field should one day become amenable to indepen-dent study (it is not currently taught formally anywhere).In addition, a journal focused on the field has beenpublished since 1997 as a joint venture of the AmericanChemical Society and the Royal Chemical Society (7).Alas, the engineering scale-up of synthetic bulk drug pro-cesses is still badly understated, as most contributors tothe new body of literature are synthetic chemists. Forcompounds derived from biosynthesis, however, there isa large body of biochemical engineering literature thatdeals in depth with the scale-up of the biosyntheses andthe subsequent ‘‘downstream processing’’ technologies(e.g., Refs. 8–10).

The application of the fruits of bulk drug process develop-ment to process design, technology transfer, and firstmanufac-ture will be addressed in the companion chapter 3, as those

Process Development 11

Page 27: Active Pharmaceutical Ingredients

activities are carried out in a distinct context that overlapswith the R&D activities. Such planes of contact will, of course,be identified in this chapter and their discussion confined tothe minimum needed herein.

About the scope of the chapter, it is ambitious in its aimto support the above objectives, yet modest in its depth ofdescriptive material, since doing justice to the latter wouldrequire a much larger volume. Instead, the author has chosento address the fundamentals along the said objectives, whilekeeping the descriptive technical material spare and aimedat selected targets of the bulk drug process development task:e.g., seeking thermochemical safety, scaling-up, achieving thedesired physicochemical attributes of the bulk drug, captur-ing and applying the process know-how.

As of this writing in 2004, the process developmentmilieu of the bulk drug industry is quite varied—from thelarge drug company in which all the skills are representedto the small virtual firm that contracts out the work, as wellas firms that do selected process development tasks as partof their attempt to secure the eventual manufacturing busi-ness from the owner of the drug candidate. The author hasnot attempted to deal separately with these different environ-ments lest the exposition of the target fundamentals getobscured by the specifics of each case. Instead, the bulk drugprocess development task is discussed within the continuumof a large drug company and commentary that applies toother contexts has been inserted, hopefully in a sparing andincisive manner.

The reader should be alerted to an additional choice ofthe author. Although the increased regulatory expectationshave deeply transformed the process development task, theparamount stance for the practitioner remains intact: knowand understand your process, reduce it to practice soundly,and operate it in a disciplined manner. Accordingly, thisand its companion chapter, aimed at the fundamentals, avoidthe spectrum of the current good manufacturing practicessubject (or cGMP), which seems to have soaked so much ofthe energy of process practitioners throughout the bulk drugindustry. However, the issues associated with the assembly of

12 Rosas

Page 28: Active Pharmaceutical Ingredients

regulatory submissions (IND, NDA and the like) and with theexpectations of the subsequent approval process will bediscussed as required to meet the objectives of the chapters.

Finally, the diligent reader of these two chapters, armedwith the perspectives provided herein, should find thatcontinued study of the literature can be quite fruitful. Toassist in that task, a selection of references is included, mostof which are cited throughout the text, with the rest citedseparately as suitable reading for the studious.

II. THE BULK DRUG PROCESS AS PART OF THEDRUG DEVELOPMENT PROGRAM

A. The Chemical Process of a Bulk Drug

In the context of this chapter, a bulk drug or a bulk drugsubstance is a material—a single chemical compound withthe desired biological activity—obtained in bulk form and des-tined for the preparation of dosage forms. The latter, whenadministered in a prescribed manner to the target patient,animal or plant, delivers the drug so as to elicit a desiredphysiological response and, in due course, the intended thera-peutic or protective result. More recently, terms such as activepharmaceutical ingredient (API) or bulk pharmaceutical chem-ical (BPC) seem to have overtaken the usage, seemingly as theresult of their adoption by regulators in the United States.Herein we will use the original term bulk drug (or bulk), as itmost aptly describes the material—a drug that is obtainedand characterized in bulk form. However, we will confine ourscope to those compounds commonly known as chemical enti-ties—drugs of relatively small molecular weight that can becharacterizedwell by currentmethods of chemical and physico-chemical analysis. Indoing so,weareexcluding thosemacromo-lecules, substances, andpreparations of biosynthetic origin thatare collectively known as biologicals. The processing methodsused in biologicals, albeit based on the same fundamentals,are significantly different from those applied to chemical enti-ties, and their process development, registration, andmanufac-ture also take place in a rather different environment. In

Process Development 13

Page 29: Active Pharmaceutical Ingredients

addition, organic compounds categorized as nutritionals andfine chemicals at large are not within this scope, their proces-sing similarities with bulk drugs notwithstanding.

Bulk drugs are obtained through three chemical proces-sing routes:

a. Extraction, recovery, and purification of the drugfrom biomasses of natural origin or from fermentation(Fig. 1): (1) paclitaxel is extracted from various Taxusplants; (2) lovastatin is biosynthesized in the fermen-tation of nutrients by Aspergillus terreus.

b. Semisynthesis, in which a precursor compoundfrom a natural source or a fermentation is convertedto the target drug by synthetic chemical modifica-tion: (1) fermentation penicillin G is converted to6-aminopenicillanic acid, which in turn is reactedwith an acyl chloride to afford ampicillin; (2) naturalmorphine is methylated to codeine (Fig. 2).

Both routes to bulk drugs take advantage of the diversityand richness of molecular structures found in naturalsources, where many important biological activities arealso found.

Figure 1 Bulk drugs from natural sources: Paclitaxel (antileuke-mic and antitumor) and lovastatin (inhibitor of cholesterol bio-synthesis) are examples of the diverse and complex structuresmade by plant and microbial cell biosyntheses, respectively. In mostinstances of such compounds having desirable biological activities,their structural and chiral complexities make chemical synthesisnot competitive with isolation from biosynthesis.

14 Rosas

Page 30: Active Pharmaceutical Ingredients

c. Total synthesis from simple starting materials orless simple intermediate compounds (Fig. 3): (1)fosfomycin from commodity chemicals, (2) lobetalolfrom 5-bromoacetyl salicylamide.

In either total synthesis or semisynthesis processing,sometimes a desired synthetic transformation is bestdone by an enzyme. Such synthesis step, whether usinga preparation of the enzyme or the host microorganism,will be considered a chemical synthesis step (a biotrans-formation or a biocatalytic step) and not a fermentationfor biosynthesis.

Whichever of these routes is used to obtain a bulk drug consti-tutes the chemical process. Further processing of the bulkdrug to obtain the dosage form constitutes the pharmaceuti-cal process. This distinction is depicted in Fig. 4, where simple

Figure 2 Semisynthetic bulk drugs: Ampicillin (antibacterial)from penicillin G. Modifications of biosynthetic structures are oftencreated to improve the in vivo attributes of the original compound,utilizing the biosynthesis product as the starting material contain-ing most, if not all, of the structural complexity that provides thebasic biological activity. Similarly, codeine (analgesic), althoughfound in opium from Papaver plants, is most economically madeby methylation of morphine, which is more efficiently isolated fromopium.

Process Development 15

Page 31: Active Pharmaceutical Ingredients

graphical means are used in an attempt to differentiate thebulk character of the product of the chemical process, in con-trast to the discrete character of the product of the pharma-ceutical (or dosage form or secondary manufacturing)process. The distinction also reflects their very different tech-nology, manufacturing, and regulatory environments.

In the current pharmaceutical parlance, the term API isused most often as descriptive of the biological activitycontribution. Herein, however, the term bulk drug isused instead as descriptive of the physical and chemicalcharacter of the subject material, with its biological activ-ity taken as obvious. Indeed, the conventional term for

Figure 3 Drugs by total synthesis: Fosfomycin (antibacterial) is agood example of the manufacture of a bulk drug by total synthesisfrom basic chemicals, albeit the compound is of biosynthesis origin.Alternatively, and more frequently, the manufacturing process issimplified by tapping on commercially available compounds ofgreater structural complexity (intermediates), such as 5-bromoacetylsalicylamide as the startingmaterial for lobetalol (antihypertensive).Even if the intermediate is custom made by others, the processdevelopment and manufacturing task for the drug developer isgreatly simplified relative to the use of basic or building blockchemicals.

16 Rosas

Page 32: Active Pharmaceutical Ingredients

the other ingredients added to formulate the dosage formis still inactive pharmaceutical ingredients.

As we proceed, unavoidably some other terms will be usedthat may not be familiar to all readers. Accordingly, aneffort will be made to define such terms at the point of firstuse, as well as to use them sparsely. For example, unitoperations are those methods that can be found repeatedlyused in chemical processing and that have a commonphenomena root, their many variations notwithstand-ing—filtration to separate solids from an accompanyingliquid, distillation to separate volatile components from amixture, or milling to reduce the particle size of particu-late solids. The organization of chemical processing onthe basis of such unit operations was crucial to thedevelopment of organic chemical technology, which was

Figure 4 The domains of chemical (bulk drug) and pharmaceuti-cal (dosage form) processing, with the chemical processing domaindefined by the shaded area of the diagram.

Process Development 17

Page 33: Active Pharmaceutical Ingredients

originally arranged on the chemistry basis of unit pro-cesses, such as nitration, sulfonation, or esterification.Whereas the latter organized knowledge on a strictlydescriptive basis, the unit operations approachmade possi-ble the study of processing phenomena on the basis of gen-eralized principles from physics, chemistry, kinetics, andthermodynamics, which could then be used to undergirdmethods applicable in the context of any chemical processand over a wide range of scale and circumstances. Hence,the keystone role that unit operations played in the adventof chemical engineering as a discipline, with a practicequite distinct from that of the earlier industrial chemistry.

B. A Perspective

Process development of a bulk drug consists of three distincttasks:

a. Preparation of bulk drug as needed by the overalldevelopment effort—the preparative task. The scopeof this task varies over a wide range, as shown inTable 1.

b. Definition and achievement of the desired physico-chemical attributes of the bulk drug as needed bythe dosage form development—the bulk drug defini-tion task.

c. Acquisition and organization of a body of knowledgethat describes a sound process for regulatory submis-sions and technology transfer to first manufacture atscale—the body of knowledge task.

However, these tasks cannot be directed to successfuland timely completion unless viewed and managed as a veri-table trinity, their differing demands and instantaneousurgencies notwithstanding. Drug development is a fast pacedand difficult enterprise; it presents frequent junctures atwhich the need to focus on the most compelling task needsto be artfully balanced with other needs lest the aggregatetask be compromised—all three tasks need to be completed

18 Rosas

Page 34: Active Pharmaceutical Ingredients

Table

1Bulk

Dru

gDem

andsof

theVariou

sDru

gDev

elop

men

tPhases

Preclinicalphase—

Initialtoxicolog

y,probes

ondru

gbioavailability,

data

gatheringfortheIN

D,additionalanim

alstudies,

etc.

Supplies

tobedelivered

over

2–6mon

ths

Total�5–50kg

Phase

I—Use

inhumans(20–80mostlyhea

lthy

subjects)

forpharm

acokinetic,pharm

acological,routesof

administration

,doseranging,andtolerance

studies.

Con

tinuing

toxicolog

yanddosageform

dev

elop

men

t.Allaim

edatthedesign

ofPhase

II=IIIstudiesanddefi

ningthetarget

dosageform

s

Supplies

tobedelivered

over

6–12mon

ths

Total�20–100kg

Phase

II=III—

Increa

singly

largenumber

ofpatien

ts(upto

thou

sands)

instudiesfortherapeu

ticeffectiven

ess(initialandconfirm

atory),

doseandregim

endetermination

,ev

aluation

oftarget

pop

ulation

sforsa

fety

andefficacy,su

pportof

desired

claim

s,mark

et-specific

anddosageform

-specificstudies,

etc.

Con

tinuingtoxicolog

yand

dosageform

dev

elop

men

t,stabilitystudies.

Allaim

edatthe

assem

bly

ofthedossier

Supplies

tobedelivered

over

18–48mon

ths

Total�300to

>2000kg.

Phase

IV—

Postapprovalstudiesforop

timization

ofdru

guse,

pharm

acoecon

omic

data,morbidityandmortality

data,

hea

d-to-hea

dandconcomitantdru

guses,

etc.

Thesestudiesare

gen

erallysu

pplied

from

bulk

dru

gmadein

the

manufacturingop

eration

.

Notes:

1.IN

D(investigation

alnew

dru

g)is

thesu

bmission

requestingUSFDA’s

exem

ption

from

dru

gsh

ippingin

interstate

commerce,thus

signalingtheintentto

initiate

studyin

humans(ortarge t

speciesifaveterinary

dru

g).Dossier

isaterm

oftenusedto

describethetotal

bod

yof

know

ledgeon

thedru

gcandidate,from

whichindividualsu

bmission

sare

assem

bledforfilingwiththevariou

sagen

cies;e.g.,the

new

dru

gapplication

(NDA)to

theUSFDA.

2.Theranges

ofbulk

dru

gtotals

reflectthewidedifferencesamon

gdru

gcandidatesandtheirprograms.Issu

essu

chasdru

gpoten

cyand

dosageregim

ens,low

anim

altoxicity,length

oftrea

tmen

tto

theclinicalen

dpoint,relativedifficu

ltyof

dosageform

dev

elop

men

t,number

ofdosageform

sdev

elop

edandscop

eof

theclinicalstudiesare

theprincipalfactorsdeterminingthedem

andsforbulk

dru

g.Obviously,

relativelyinfreq

uen

tex

trem

esex

iston

both

ends:

from

alow

end

fordru

gssu

chasdizocilpine,

paclitaxel,and

someex

perim

ental

oligon

ucleotides

toahighen

dforHIV

protease

inhibitors(highdoses)andsomecard

iovasculardru

gs(clinicalstudiesof

verylargescop

e).

Sou

rce:

Author’sob

servation

sfrom

involvem

entin

numerou

sdru

gdev

elop

men

tprograms.

Process Development 19

Page 35: Active Pharmaceutical Ingredients

at the same time for timely and successful product launch.Selected instances of such balancing, in which some risk isoften inevitable, are discussed throughout the rest of thechapter; therein lies the crucial need for overall coordinationof each drug’s development program.

Although various models exist, today’s drug developmentis generally facilitated by a coordination mechanism andforum, usually in the form of a cross-functional team thatdrives and manages a drug candidate. The principalobjectives are to have and execute: (a) a drug develop-ment plan, (b) rigorous means to closely track its execu-tion, and (c) mechanisms to effectively respond to eventsand findings that invariably arise in spite of the plan.Indeed, the development of a new drug encompasses amyriad activities and objectives that are extremelycross-linked among the various disciplines contributingto the effort. Clearly, the bulk process development teamneeds to be well represented in the cross-functional forumthroughout the drug development cycle.

Success in development coordination means that, nomatter which coordination model is used, there must beprompt and effective resolution of most issues and difficul-ties, say > 90%, at the team level, with the rest going up toa broader and more senior team of the R&D organization(i.e., the heads of the disciplines, functions, and thoseabove). Indeed, the direction and operation of such teamshave become a distinct function (it will be referred hereinas drug coordination), with its own set of skills and notunlike the distinct set of skills in new drug submissionsand approval—the regulatory affairs function.

The relationships of the three basic tasks with the over-all drug development program are depicted in Fig. 5 in rathersimple terms, whereas the specifics of each relationship willbe discussed under the heading of each task. The arrows indi-cate the flow of materials from the preparative task and theflow of information and know-how from each task to theothers and to the drug development at large.

It is also useful to depict the bulk drug process develop-ment cycle on a Cartesian coordinate plane (Fig. 6). The

20 Rosas

Page 36: Active Pharmaceutical Ingredients

abscissa axis represents progress since the onset of develop-ment of a compound and, although progress along well-defined milestones is used, one might also look at the abscissaas measuring the applied technical effort or, less precisely,the extent to which the bulk drug process has been reducedto practice (e.g., kilos of bulk drug made, batches made, orversions of the process piloted). Inevitably, the abscissa scaleshown herein is arbitrary, albeit deliberately selected; theexperienced reader will probably readily think of an examplewith a more apt progress scale. Thus, the need to deal withthe latter in terms of more distinct stages, which Fig. 6attempts to depict.

Were elapsed time to be used, the distance betweenPhase II=III start and the Dossier filing milestones would bequite variable from drug to drug, as that interval dependson the scope of the clinical program and on the therapeutictarget. Whereas osteoporosis, diabetes, or depression require

Figure 5 The three basic tasks of bulk drug process development.These tasks exist concurrently throughout most of the developmentcycle, albeit their burdens vary through the cycle. Nevertheless,managing well all three tasks as inseparable parts of a singleoverall endeavor is the principal managerial challenge in bulk drugprocess development.

Process Development 21

Page 37: Active Pharmaceutical Ingredients

considerable time to reach their efficacy endpoints, those forbacterial infection or pain relief, for example, arrive muchsooner. For this, and for other reasons related to the intendedscope of the drug development (e.g., claims structure, scheduleof filings, multiple routes of administration, etc.), the elapsedtime scale is unsuitable for the process know-how purposesof Fig. 6. Instead, applied effort or extent of reduction topractice of the process relate directly, if not strictly in directproportion, to the acquisition of the process know-how.

Although the biobatch and preapproval inspection prere-quisites are specific to USFDA approvals, analogous expecta-tions are arising in other drug agencies in the major markets(more on this in Chapter 3). The biobatch is a distinct markerin dosage form development in that it serves as the bioavail-ability=bioequivalence bridge to pivotal clinical studies, aswellas the bioavailability=bioequivalence reference for all subse-quent dosage form output. As such, the biobatch reflects the

Figure 6 The process know-how vs. applied effort plane, includ-ing the major milestones of bulk drug process development. Asdefined herein, 100% know-how describes the body of knowledgeneeded for registration and reliable first manufacture for productlaunch, whereas additional know-how accumulates with manufac-turing experience and follow-up work that might be done for processimprovements or a second generation process.

22 Rosas

Page 38: Active Pharmaceutical Ingredients

process that goes into the dossier,uses representative bulk drugand excipients, and its size is no less than 10% of the intendedmanufacturing scale. Preapproval inspection is a methodologyemployed by the USFDA to ascertain, at its discretion, thatthe intended manufacture of dosage form and bulk drugcorrespond to the processes used in the pivotal clinical studiesand described in the NDA or other new drug submissions.

The ordinate axis, on the other hand, is straightforward,as it measures the fractional bulk process know-how relativeto that required for regulatory approvals and for sound firstmanufacture. Note, therefore, that it is not being suggestedthat at 100% on the ordinate axis there is nothing else to belearned about the process; instead, the 100% ordinate valuemerely describes the knowledge required to fulfill the said pro-cess development objectives. Indeed, further gains in processknow-how are always realized with manufacturing experi-ence, and mature processes often differ appreciably from theirfirst manufacture versions, by virtue of gradual improve-ment or from significant step changes (second-generationprocesses), although most often the seeds for such later devel-opments are planted in the original development body ofknowledge. Thus, the curve in Fig. 6 describes the accumula-tion of know-how during four distinct phases of the processdevelopment effort:

a. The preparative stage, during which the effort isfocused on making available kilogram amounts ofthe bulk drug to the preclinical, toxicology, andPhase I work, usually not based on the eventualsynthesis route, let alone the eventual process.

Whereas the synthesis route (or scheme) describes theintermediate chemical structures sought to arrive at thefinal compound (starting materials, synthesis approach,and probable chemical reactions to use), the processdescribes how the route is implemented at a much higherlevel of detail. (solvents, catalysts, purifications, isola-tions vs. straight-through, etc.).

b. The development stage, in which the preparativework is scaled up and the synthesis effort goes into

Process Development 23

Page 39: Active Pharmaceutical Ingredients

high gear, aimed at the manufacturing route andprocess. It is in this stage that the chemical engi-neering effort is applied in earnest, first to supportthe scaled-up preparative work and then to addressthe scale-up issues of the manufacturing route.

Ideally, the chemical engineering contribution startsearly, so as to appropriately influence the seminal choicesbeing made by the process chemists as to route. Thisinfluence is reasonably apparent with respect to issuesof thermochemical safety and probable environmentalimpact; yet, there is across-the-board synergy that a che-mistry=engineering dialogue can exploit. The latter isparticularly true in those instances when the chemistsperceive a desirable approach as not being feasible ongrounds of scale-up difficulty or, more simply, becauseof lack of experience with some demanding processingconditions.

c. The consolidation stage, in which the synthesis routeis fully settled and the specific process for it is definedat the level of detail that permits process design forthe manufacturing plant, definition of the bulk drugattributes and the assembly of the dossier. Also dur-ing this phase all the preliminaries for technologytransfer are carried out and the stage set for firstmanufacture.

d. The technology transfer stage, in which the process isrun in its first manufacturing venue, its performanceestablished, and the bulk drug needed for productlaunch is produced. Also during this phase, the manu-facturing scheme receives approval within theapproval of the dossier, often after plant inspectionby the approving agencies.

From the above definitions, a discussion of the specifics ofeach stage is now possible, also based on the depiction of thebulk drug process development cycle on the know-how vs.applied effort plane introduced in Fig. 6. During thesestage-specific discussions, the three bulk development taskswill serve as the basis and along the lines of Fig. 5.

24 Rosas

Page 40: Active Pharmaceutical Ingredients

C. The Stages of Bulk Drug Process Development

1. The Preparative Stage

Although preparative work takes place throughout the pro-cess development cycle, this first stage is most aptly describedas the preparative stage. Its focus, although not exclusively, isthe preparation of limited amounts of bulk drug for assortedpreclinical purposes, then followed by first scale-up to supportPhase I activities, which include testing the drug in healthysubjects (humans or the target animals if a veterinary drug).

Starting with bench scale equipment (up to 100L in theso-called kilo lab) or pilot scale fermentors (up to 5000L whentiter is low), this early preparative work uses whatever syn-thetic method or fermentation conditions (the microorganismand the nutrients) are immediately available. In most cases ofsynthesis, the route may be a somewhat streamlined versionof the discovery route or a temporary route that may or maynot include parts of synthesis schemes being considered foreventual development. In most cases of biosynthesis, themicroorganism is that from the discovery stage, but takenfrom whatever stage of microbial strain improvement isamenable to scale up from shake flasks or bench scalefermentors.

Fermentation processes at this stage are generally ofvery low productivity (final concentrations of the targetcompound of < 1g=L), making access to relatively largefermentors most helpful, including, in cases of dire need,the use of manufacturing scale units (up to 75,000L), thepoor scaled-up performance of the early stage notwith-standing. The analogy for chemical synthesis is the ardu-ous operation of lengthy procedures in the kilo lab, thelow yields notwithstanding.

Although the kilo lab will be described more fully lateron, it may be said at this point that the kilo lab is a largerscale lab, traditionally used for running preparativeprocedures than for experimentation.

Preclinical and Phase I development work is crucialin that it determines the merit of further development or,

Process Development 25

Page 41: Active Pharmaceutical Ingredients

hopefully, the adjustments that need to be made to movethe compound forward. Thus, the importance of providingthe required material on time to get those answers as soonas possible. This reflects on the need for capital investmentin facilities such as kilo lab or pilot plant, and we will discusselsewhere in this chapter the challenges of this stage ofdevelopment when the preparative stage depends on outsour-cing (the reliance on outside suppliers). Indeed, sufficientinternal resources for the preparative stage is a clear compe-titive advantage, with the optimal setting providing themeans—hardware and engineering skills—to swiftly overlapthe kilo lab work with pilot plant work-up to, say, 1000Lvessels and the appropriate auxiliaries and operating envir-onment (safety, industrial hygiene, and pollution abatement).Figure 7 depicts this preparative environment, whereas

Figure 7 The resources for the preparative task. The need toengage larger-scale resources depends on the scope of the prepara-tive task, which can vary widely (Table 1 and Fig. 8).

26 Rosas

Page 42: Active Pharmaceutical Ingredients

Fig. 8 complements the range of preparative scopes pre-sented in Table 1.

Also depending on the resources of the organization,synthesis bench work may take place in search of routes thatcan support a manufacturing process, as the routes used dur-ing the discovery phase are largely unsuitable on the basis ofprojected cost, length of the synthesis cycle, commercial una-vailability of starting materials or simply because of their per-ceived inferiority relative to what the process chemists foreseeas attractive alternatives. Clearly, the compelling wisdom ofsuch early synthesis work needs to be balanced against theresources available and, most of all, against the empiricalprobability of less than 20% that a drug candidate at thatstage will reach the market, as indicated by Table 2.

Whereas medicinal chemists practice organic synthesis asan indispensable tool and are largely oriented upstream(towards the domain of biological and pharmaceuticalattributes of the compounds they work with), process

Figure 8 The scope of the preparative task. Some examples toillustrate the dependence of the preparative effort on drug potency,therapeutic target, and scope of the clinical effort.

Process Development 27

Page 43: Active Pharmaceutical Ingredients

chemists in the drug industry practice synthetic chemis-try as their profession and are oriented downstream(towards the reduction to practice beyond their labbench), thus the usual discontinuity in synthetic routeat the discovery=development boundary.

Although sometimes much is made about smoothingand simplifying the discovery synthetic route (eliminatingisolations and purification, shortening the processing cycle,and using less expensive materials), the most desirablecontribution of the process chemist is the conception of adistinctly advantageous synthesis route that can then bedeveloped and engineered into a sound manufacturing pro-cess. Such a route would bring the advantages of fewer stepsfrom reasonably available starting materials, environmentalbenevolence (or, preferably, green chemistry), parallel moi-eties that can converge into shorter synthesis cycles, stereose-lectivity, and similarly decisive gains.

As a summary, Fig. 9 focuses on the preparative stageand the rest of the preparative effort on the know-how vs.applied effort plane, whereas Fig. 10 depicts the materialsflow from the bulk drug preparative effort at large.

Table 2 Best Practices Probabilities of a Drug CandidateReaching the Market

Drug candidates in the preclinical phase 5–10%In Phase I 10–20%In Phase II 30–60%In Phase III 60–80%Post NDA filing > 95%

Notes: ‘‘Best practices’’ refers to drug development organizations with establishedgood records of bringing drugs to market. In particular, best practices include a highhurdle for a drug candidate to enter development or Phase I.Source: Author’s assessment from assorted estimates, including those from thePhRMA Annual Report—online edition, 1997. While the figures from totalcompounds synthesized (or total number of biologically active compounds) haveincreased as the methods for generating actives improve their total output, the abovefigures after entry into development have remained largely unchanged. The aboveranges probably reflect the adequacy of the tools used to assess the merit ofdeveloping an active compound and the rigor of the criteria for moving a compoundforward.

28 Rosas

Page 44: Active Pharmaceutical Ingredients

Figure 9 The preparative effort in the know-how vs. appliedeffort plane. The principal preparative milestones are shown.

Figure 10 Materials flow from the bulk drug preparative effort.The width of the arrows approximately indicates the relativeamounts of bulk drug going to the users in the overall drug develop-ment program. Examination of this figure and Fig. 9 provides anequally approximate description of the bulk drug usage as a func-tion of the development cycle.

Process Development 29

Page 45: Active Pharmaceutical Ingredients

2. The Development Stage

As made clear by the slope of the curve in the know-how vs.applied effort plane (Fig. 6), the development stage comprisesthe most productive development effort:

(a) Synthesis work at the bench scale seeks the even-tual manufacturing route in earnest, preferably on morethan one approach, with all promising a substantial, ifnot overwhelming, advantage over the current preparativeprocedures.

In chemical synthesis, the route is basically driven by thestructure of the target compound. Within that logic, however,the creativity of the process chemist is bounded only by therealities of starting materials availability. However, examplesof bulk drugs made from commodity chemicals are now fewand rapidly disappearing (thiabendazole and l-methyldopa,for example), as the more complex structures of today’s med-icinal chemistry preclude synthesis from basic raw materials.Instead, today’s process chemist must be very alert to whatthe fine chemicals industry offers (or could be induced to offer)by way of suitable building blocks or intermediates and thecorresponding manufacturing capabilities. Such alertness,combined with creative synthesis skills, is the key to trulyadvantageous routes. This theme is discussed amply and indepth in some of the previous references (2–5), as well as inSaunders’s (11) compendium of selected major drugs. In theextreme, the total synthesis of structurally rich naturalproducts, although rarely aimed at a manufacturing process,offers leads and inspiration to the process chemist, as wellas blazes the trail with new reactions, some of which areeventually used in bulk drug syntheses (e.g., Ref. 12).

In celebrating the opportunities for the creative processchemist, we should not neglect factors such as the increas-ing desire for environmentally benevolent chemistry(green chemistry) or the prevailing business model in thebulk drug industry, by which the range and scope ofchemical processing has been narrowed in favor of con-tracting out (outsourcing). There is also, onmanagement’spart, the reluctance to practice hazardous chemistry

30 Rosas

Page 46: Active Pharmaceutical Ingredients

(nitration, sulfonation, phosgenation, etc.), with thatspectrum of processing now all but ceded to contractmanufacturers.

Some compounds of natural origin products have beenmanufactured by total synthesis when structurallysimple (e.g., chloroamphenicol, fosfomycin) or wheninevitable to bring a significant drug to market, as inthe case of imipenem (13).

The selection of the chemical route, which is invariablymade before it has been sufficiently reduced to practice, isthe strategic decision, as it has the greatest potential to definethe process and its overall performance—costs, reliability,environmental impact, etc. Accordingly, it is a decision thatis best made with the benefit of sufficient engineering assess-ment, as sometimes the chemical appeal is not sufficient.Indeed, engineering assessments of capital and operatingcosts, environmental impact and issues of process designand scale-up bring sharply into focus the general directionas well as the specific development actions that the routerequires to become the manufacturing process. On occasion,such assessments cause reappraisal of the route that, iftimely, can redirect the project to considerable advantage—to a superior variation within the same basic route, or to asubstantial change to a hybrid chemical scheme and, lessfrequently, to abandonment for another route.

Preferably, the synthesis route is settled not late duringthis stage, but it is not all that rare, in the higher caliberprocess efforts, for that ‘‘better route’’ to come throughand displace the prevailing route just in time to switchthe scaled-up preparative work.

It is at this stage of merging chemistry and engineeringefforts that the process development effort generally settlesonto the right track and the effort approaches critical mass.Process development organizations that lack the requisiteengineering skills or that tap into relatively distant skills(say, from a technical resource in manufacturing) are at amarked disadvantage with respect to choosing the betterprocess, since the said assessments are not done, are done less

Process Development 31

Page 47: Active Pharmaceutical Ingredients

effectively, or done without the criticality of mass that theoccasion demands. The distant engineering skills are alsofar less persuasive when their assessment of the proposedsynthesis is not favorable.

All seasoned practitioners of bulk drug process develop-ment know from at least one experience the very high pricepaid when the wrong process gets too far down the develop-ment cycle, and retreat is either unacceptable or very costlyto the overall development timetable. Thus, the compellingneed to make the fundamental choices of route, and of processapproach within the route, with the full set of skills andaddress the key questions:

1. What will the commercial plant look like? What willits operation be like?

2. What are the probable capitals costs? How long willit take to be ready to start up?

3. What are the scale-up issues? Can they be addressedon time?

4. What is the environmental impact? Is there a goodfit with the likely plant sites?

Once the bulk process team gets past this juncture withan action plan, the rest of the development stage is mostly amatter of good execution by all the disciplines involved.Although the Analytical R&D function has not been men-tioned up to now, its role is, of course, pervasive throughout;first in support of the early preparative work (a duty thatremains with the function for the rest of the developmentcycle), then in decisive and indispensable participation ofthe development activity at the bench and in the pilot plant.

Biosynthesis processes, which are based on fermentationprocessing inwhich themicroorganismdoes the synthesis, facethe same set of development issues, but in a narrower field ofoptions. Not only is the biosynthesis well defined and fixedby the microorganism, but alternate microorganisms withradically different pathways that could be more desirable arenot that available. Chemical entities of natural originare secondary metabolites of microorganisms or plant cells,and variations in the metabolic pathways that lead to a

32 Rosas

Page 48: Active Pharmaceutical Ingredients

given secondary metabolite are relatively narrow compared tothe many variations by which a compound can bemade by che-mical synthesis.

In this case, the development team (microbiology andbiochemical engineering) aims at coaxing the organism orplant cell to be more effective. Strain mutation is a proventechnique for improvement of the productivity of microbialbiosynthesis. Plant cell processes, although very few in indus-trial practice, also seem amenable to increased productivityby manipulation of the cell lines and fermentation conditions.The microbiologist and the biochemical engineer are thus ableto offer the potential for increased fermentation output by fac-tors up to an order of magnitude or more—a potential not tobe matched by increased yields from an organic synthesis.Indeed, some fermentation processes can go into manufactureat low titers with a high probability that increases will beobtained with continued development of the microbe or plantcell, as well as the fermentation conditions. Thus, variationson the biosynthesis—unlike variations on how to chemicallysynthesize a compound—are modest in range, but not insignificance to fermentation productivity (e.g., use of phenyla-cetic acid as a precursor in the fermentation of penicillin G) orother important aspect of the process (e.g., switching to a dif-ferent Taxus plant from which a precursor to paclitaxel,comprising the taxane ring with all of the desired stereochem-istry, could be extracted and chemically converted topaclitaxel at an advantage over the prior extraction ofpaclitaxel).

It is in the processing downstream of the fermentor thatdevelopment possibilities become numerous, as a wide rangeof unit operations for concentration, purification, and isola-tion exists, just as wide as the processing options for recover-ing the desired compounds from streams (i.e., materials)issuing from chemical synthesis. This is discussed muchfurther elsewhere in this chapter.

(b) It is also in the development stage that the prepara-tive work is scaled up in earnest with two purposes: (1)greater output of bulk drug and (2) the identification andresolution of the problems of scale attendant to the desired

Process Development 33

Page 49: Active Pharmaceutical Ingredients

process. Although the latter goal requires that the desiredroute be at the scaled-up stage, considerable progress can bemade if pieces of the desired route are scaled-up before thetotal route is brought to the pilot plant.

(c) It is also during the development scale that the defi-nition and achievement of the desired physicochemical attri-butes of the bulk drug is pursued in earnest, hopefully afterthe dosage form development team has narrowed down theranges for those properties after the major decision—whichparticular salt or the free base or the acid will be the bulkdrug form of the biologically active structure. Such a decisionmay come late in the cycle, for oral drugs in particular, as thesearch for the desired bioavailability and stability may bearduous (14).

(d) Finally, it is during the development stage, prefer-ably early, that the bulk development team starts its workwith the appropriate downstream organization in anticipa-tion of successful drug development, registration, and marketlaunch. This set of activities takes place in a rather distincttrack from the R&D track, often placing inordinate demandson the bulk process team, as their obligations to the drugdevelopment effort remain unaltered by the onset of theirobligations to eventual technology transfer.

There is a great deal of risk when bulk process resourcesare badly caught in the vise of the demands from theirdrug development partners and the increasing demandsof technology transfer. Staffing of the bulk process team—the engineers in particular—needs to recognize thatsuccessful drug development brings with it technologytransfer. Unfortunately, R&Dmanagement and the peersin the drug development program are often insensitive oroblivious to the situation and the cross-functional coordi-nation team needs to be indoctrinated accordingly. It isvery helpful to have the downstream functions relatedto manufacturing participate in the coordination teamand thus ensure that those demands get known, if notfully appreciated.

In summary, Fig. 11 depicts the development stage in thenow familiar know-how vs. applied effort plane. It is also

34 Rosas

Page 50: Active Pharmaceutical Ingredients

timely to present the full spectrum of the bulk drug develop-ment disciplines and all the activities that they carry out,including those shared with others in the corporation or withoutsources, as shown in Fig. 12.

3. The Consolidation Stage

Although it is not infrequent for a significant bulk process‘‘loose end’’ to remain tenaciously loose until late in the cycle,by and large the development cycle reaches a stage at whichthe more difficult development work has been done. To wit:

a. The chemical synthesis route is fully defined, albeitsources and specifications of starting materials maystill be under negotiation or definition.

b. The actual process based on the synthesis route issufficiently defined and sound pilot plant operatingprocedures exist or are clearly in the offing.

c. Preparative support to the drug developmentprogram, although continuing and never leisurely,

Figure 11 The development stage in the know-how vs. appliedeffort plane. The principal process development milestones areshown.

Process Development 35

Page 51: Active Pharmaceutical Ingredients

is no longer threatened by uncertainties about howto prepare the bulk drug.

d. Thermochemical safety data are firm and only updat-ing for process changes remain to be done. All issuesare being dealt adequately in the process design ofthe manufacturing plant.

e. The environmental impact of the process at the site ofmanufacture and at large is understood and accepta-ble, meeting company policy objectives. Obtaining allthe requisite permits is likely.

f. Industrial hygiene issues specific to the process areunderstood and being addressed adequately in theprocess design of the manufacturing plant.

g. The process design, and possibly plant construction,are proceeding. Uncertainties seen within the graspof the combined development=process design effortand work can be focused accordingly.

Figure 12 Disciplines and activities in bulk drug process develop-ment. CMC stands for the chemistry, manufacturing and controlcomponent of the dossier.

36 Rosas

Page 52: Active Pharmaceutical Ingredients

h. Analytical methods for in-process and bulk drug con-trol have been largely defined and remain to be con-firmed and validated. Absolute purity, impu-rity profile, and crystal form are settled matters.

i. The scope and approaches to the dossier are largelyin hand, if not in text.

There is, of course, no suggestion of the work being com-pleted. Far from it, the consolidation stage is intense in a dif-ferent way that the development stage was. A great deal of thework ahead is filling blanks (few if the prior work has beendone well), refining pilot plant procedures and catching upon the documentation that will support the dossier. Also,the final work on the definition and achievement of the bulkattributes needs to be done to support the final work on thedosage form side and the biobatch and stability studies thatwill follow.

There is also the largely increased workload in prepara-tion for technology transfer, usually requiring frequenttravel, a great deal of interaction, and the pursuit of muchdetail. Snags in process design and plant construction docome up, environmental permits may require scrambling forsome data, etc.

However, the slope of the know-how curve is decreasingrapidly, as the bulk process is being implemented more thanit is being developed, the loose ends notwithstanding. In sum-mary, Fig. 13 depicts the consolidation phase in the know-howvs. applied effort plane.

4. The Technology Transfer Stage

Most of the discussion on the nature and scope of the technol-ogy transfer activity is presented in Chapter 3. Nevertheless,the following seems pertinent at this point, as it relates tothe technology transfer burden that the bulk process deve-lopment team carries in addition to its duties on the drugdevelopment program:

a. A finite effort, even if the midst of a very difficultdevelopment stage, must be allocated to look ahead

Process Development 37

Page 53: Active Pharmaceutical Ingredients

to the specifics of manufacturing the bulk drug. Thishas been indicated in Figs. 11 and 13 .

b. The bulk process team needs to keep the rest of theR&D organization, their peers in the coordinationteam in particular, aware of this downstream task.

c. The technology transfer team needs to be wellrounded—chemists or microbiologists, engineersand analysts—and at the site of technology transfer.Staffing and briefs to do the job should be generousto decisively start up the process for product launch.No rescue missions allowed!

d. Successful technology transfer—from early planningfor manufacture, process, and plant design, processstart-up preliminaries and the actual demonstrationthat the process works in the commercial plant—rests squarely on the process body of knowledgebeing as complete as needed by the task and orga-nized to effectively impart knowledge to the down-stream organization.

Figure 13 The consolidation stage in the know-how vs. appliedeffort plane.

38 Rosas

Page 54: Active Pharmaceutical Ingredients

e. Regardless of what organizational arrangementmight exist, the bulk process development teamneedsto assume, hopefully in a collaborative understand-ing, a leadership role as the bringers of the know-how.

f. With the necessary adjustments, all of the aboveapply when transferring the process technology tocontract manufacturers or licensees. More on thisunder VI—Outsourcing in bulk drug processing.

III. FROM THE BENCH TO THE PILOT PLANTAND BEYOND

A. Process Conception and Bench ScaleDevelopment

Except for fermentation or recovery from natural sources, allother chemical entities are obtained by chemical synthesisfrom organic chemicals and the process conception starts withthat of the synthesis route—the scheme by which selectedstarting structures are converted to the target drugcandidate. Factors considered by the synthetic chemistryteam are:

(a) Starting materials that are available (or could beavailable) that promise an attractive route, and a wish listfor such a route could be as follows:

1. The route is direct, with few steps needed to reachthe target compound.

2. It is also convergent (two moieties can be assembledin parallel, then joined near or at the target com-pound), thus offering shorter synthesis cycles andhigher yields.

3. If chirality is sought, it appears attainable throughenantioselective methods.

4. Once chirality is obtained, the route preserved it.5. Minimal need for blocking=deblocking.6. Absence of highly hazardous materials, reactions, or

intermediates.7. Environmentally benevolent (i.e., green chemistry).

Process Development 39

Page 55: Active Pharmaceutical Ingredients

8. Probable cost is appropriate to the product.9. Fits nicely with existing plant running a related

process.

The relative priorities of these factors vary widely, asthey are seldom all present; neither are they fully indepen-dent from each other. For example, directness of synthesismay come at the price of a very expensive reactant, or wouldrequire that a very hazardous intermediate be made and per-haps isolated. Or perhaps the greenest route seems least fea-sible. Additionally, the selection may be constrained bycompelling demands of the drug development program: forexample, the most attractive route would take longer to beready for preparative work and development; it has to deferto the lesser route that can prepare bulk drug now—not anuncommon juncture and decision, although it can be subse-quently reversed.

Indeed, there is no established system to deliver the bestor even a very good choice of synthesis route, and creativityand synthesis acumen still dominate, although obviouslyaided by the above and other simpler criteria, such as thatof ‘‘atom economy’’ (how many atoms of the reactants end inthe final compound?) (15). Occasionally, the choice is facili-tated by a compelling case of an ideal starting material avail-ability (e.g., a chiral intermediate that would bring all or agood deal of the target chirality with it), a selling approachthat fine chemical producers exploit. Then at some point soon,the leading choice of route needs to be challenged by thevarious engineering assessments described in II. C.2.a.

Bench development of the route (or routes) of choice ispursued aggressively, ideally by both synthesis chemistsand chemical engineers, with the former elucidating reactionpathways and byproducts, seeking superior reaction condi-tions (solvents, catalysts, auxiliary chemicals, temperature,pressure, concentrations, reactant ratios, and approximatekinetics) as well as probing work-up and isolation methods.The engineers work, in collaboration with the chemists, onaspects of the chemistry better suited to their skills (e.g.,kinetics and thermochemistry, multiphasic reaction systems

40 Rosas

Page 56: Active Pharmaceutical Ingredients

with mass transport effects that distort the chemistry, or veryfast reaction with selectivity issues that are sensitive tomixing, or reactions requiring concurrent separation or contin-uous reactors with tight control of residence time or extraor-dinary heat removal provisions, etc.).

Such bench development by both disciplines is whattransforms a synthesis route into a process candidate forscale-up and eventual manufacture. If done concurrently—as it should be—it allows for the results to flow across thedisciplinary boundary and shortening the path to a soundprocess derived from a sound choice of route.

(b) Fermentation or natural product extraction pro-cesses, on the other hand, are not burdened by a broad rangeof route possibilities, as discussed under II.C.2.a. Benchdevelopment by microbiologists and engineers, however, isindeed rich with possibilities. To wit:

For microbial or plant cell fermentations:

1. elucidation of the pathway to the secondary metabo-lite;

2. nutrient, precursors, and optimization of fermenta-tion cycle conditions (from the above results);

3. strain and cell line improvements with respect toproductivity and robustness in fermentation;

4. data gathering to support scale-up to stirred tanksat all pilot plant scales;

5. definition of the downstream process candidate forrecovery, concentration, purification, and isolationof the target product from the fermentation.

For extraction of compounds from natural sources (plantor animal material):

1. evaluation of differing sources of the compoundbearing materials;

2. pretreatment conditions for successful extrac-tion;

3. extraction or leaching conditions, solvent, orextracting stream (i.e., material) selection, andseparation of spent plant material;

Process Development 41

Page 57: Active Pharmaceutical Ingredients

4. definition of the process candidate for concen-tration, purification, and isolation;

5. data gathering to support scale-up.

Most likely, both technologies eventually have to dealwith relatively large volumes of cell mass or plant materialwaste, and bench work to address those issues is also needed.

B. Process Scale-Up

1. What Is Scale-Up?

At its simplest, scale-up is the set of processing issues thatarise when the same operations take longer to execute inlarger scale equipment than at the bench scale. Althoughsuch issues do arise, they can be anticipated and in most casesavoided or largely mitigated through changes to the designand operation at the larger scale.

Much more often and less apparent, however, are theprocessing issues created by operating at larger scale—withgreater dimensions and different geometries—and thus affect-ing flow regimes, phase separation rates, interfacial surfaceareas, mass and heat transfer rates, flow patterns, heteroge-neity in process streams (i.e., materials) and many otherdimensionally sensitive variables and parameters. Theseeffects are not related to a different time scale of processingevents, but arise instead from strictly physical effects thatdistort the process results from those at the small-scale base-line, including chemical outcomes. Relevant examples are:

a. Reactants to a system of fast reactions cannot bemixed fast enough and fractions of the reaction massproceed for finite times at concentrations very differ-ent from the intended average concentration (somefractions unduly rich in the reactant being added,while others are unduly low), resulting in a productdistribution different from that predicted by thekinetics or obtained at the smaller scale.

b. Mixing in larger stirred tanks, if not adjusted prop-erly, can result in significant differences in thecomposition of matter of multiphase process masses

42 Rosas

Page 58: Active Pharmaceutical Ingredients

across the tank volume relative to the more uniformresults in smaller tanks.

c. Rotating devices of larger diameter, such as agitatorsand pump impellers, as well as internal moving partsin a solids mill, will exhibit higher tip linear veloci-ties and thus generate greater shear stresses influids or contribute greater energy to impacts rela-tive to the analogous operation at the smaller scale.

d. Crystallization processes at larger scale can sufferfrom unwanted nucleation as the result of hetero-geneities in solvent phase composition during semi-batch addition or in local temperatures upon cooling,as well as more prone to crystal attrition and contactnucleation from the higher tip speed of the agitatorsand greater energy impacts among particles.

e. Transfer rates of sparingly soluble gases into liquidsin stirred tanks generally suffer with increasingscale of the tank unless provisions are made tomitigate the differences, as the gas bubble size dis-tribution (and with it the interfacial surface area)generated by the agitator impeller is different.Hydrogenation rates observed in laboratory pressurevessels, for example, most often do not scale up topilot scale stirred tanks because of the extraordinarygas absorption obtained in the liquid vortex at the labscale; the larger pilot scale tank, being equipped withbaffles, does not generate a vortex and that contribu-tion to gas absorption is not present.

f. Large process vessels lose heat less rapidly thansmaller vessels at the same internal and ambient con-ditions and, when deliberately cooled, will cool lesseffectively, absent a mitigating cooling provision.

g. Larger continuous contacting vessels for devicesfor gas=liquid, vapor=liquid, solid=liquid, and liquid=liquid systems will perform less well because of mal-distribution and bypassing of the phases worsens asthe cross-sectional area of the contacting vesselincreases. Such scale-up requires that provisions bemadewith internal parts to alleviatemaldistribution.

Process Development 43

Page 59: Active Pharmaceutical Ingredients

h. Flow vessels will exhibit different flow patterns andresidence time distributions than smaller vessels,which need to be taken into account so as to designthe larger vessel accordingly.

Indeed, carrying out a processing operation at a suffi-ciently larger scale often shifts the rate controlling step ofthe process event from one domain to another. As an example,in reactions in gas=liquid systems the small scale usually per-mits the reactant in the gas phase to be abundantly availableto the liquid phase (the rate of chemical kinetics is observed,as the gas=liquid mass transfer is not limiting); whereas uponscaling up the gas=liquid transfer may become limiting andthe reaction, now starved for the reactant being supplied bythe gas phase, does not follow its expected kinetics. The resultof such shifts may go beyond the different rates of reaction, asselectivity (and relative rates of impurities formation) maychange upon lack of a reactant. Generally, chemical reactionsystems that have very fast rates or that take place in multi-phase systems are sensitive to the operating scale due to theintrusion of mass transfer effects upon the performance of thechemical kinetics.

The above is a partial list of frequent scale-up issues thatarise in bulk drug processing with consequences of lowerchemical yields, or worse yet, loss of control over the impurityprofile, as well as slower processing, excessive damage tomicrobial cells and crystalline solids, undesirable particle sizedistributions and any from a wide range of assorted shortfallsin process performance.

Understanding, predicting, and dealing with these issuesrequires more than a modicum of chemical engineering skills,such as fluid mechanics, mass and heat transport, the use ofdimensional analysis tools, and mathematical methods for thesimulation of events in a new context. Absent those skills,scaling-up will result in surprises, cause much less effectivetrouble-shooting, and engender an unwarranted fear of scal-ing-up. Indeed, such apprehensions are now codified in arbi-trary batch size ratios beyond which regulatory constraintsto process change apply.

44 Rosas

Page 60: Active Pharmaceutical Ingredients

Often enough scale-up is done much too tentatively,inserting intermediate scales that are not needed. Directscale-up from the lab to the plant is quite feasible in anumber of cases (e.g., fast liquid phase reactions withknown kinetics and thermochemistry). All that isrequired is that the issues be understood and the properparameters reproduced or improved at the large scale,using adjusted process conditions, as it is the set of thedefining parameters what needs to be reproduced, notnecessarily each process condition.

Failure to understand scale-up issues equates a change inscale with a change in the process. While it is appropriatefor a change in operating scale to come under the scrutinyof a well-managed change control system, there should beno assumption that it is ‘‘the process’’ that is being chan-ged; a distinction that is not about semantics, but aboutthe approach to scale-up by the practitioner. This pertainsin particular to operation of a pilot plant, in which scalingup and changing the process are a daily overlap that, ifnot practiced with a sufficient understanding of what ishappening, will often befuddle the practitioner.

Yet, scale-up is inevitable, even in the relatively lowthroughput environment of bulk drugs. Skillful use of thepilot plant environment, by which the preparative task andthe process development scale-up coincide in time and place,is essential to a vigorous bulk development program lest theactivity oscillate between the extremes of unskilled scale-upand fear of scale-up. Indeed, lack of sufficient scale-up skillsis a major disadvantage in bulk drug process development.

2. Tools for Scaling Up

In addition to the engineering skills and the access to the fullrange of supporting laboratory capabilities (bench develop-ment, in-process, analytical, physical chemistry,microbiology),scaling-up requires a variety of measurement apparatus (e.g.,compressibility cell to measure flows through beds of solids atdifferent compression), as well as the frequent assembly ofdedicated apparatus or pilot units (e.g., units to measurefouling rates of surfaces over short-term test, small-scale

Process Development 45

Page 61: Active Pharmaceutical Ingredients

centrifuges to more reliable measure centrifugation rates, leaftest units for vacuum filtration tests). It so happens that oftenenough studies and measurements cannot be made in proces-sing equipment nearly as well as they can be made in a smal-ler-scale apparatus dedicated for the purpose at hand. Theenterprising scale-up team will, in due course, assemble andaccumulate such test apparatus as the needs arise.

In addition, some scale-up works need apparatus thatare operated for preparative purposes as well, along the linesof the kilo lab, but in a flexible environment not focused exclu-sively on batch processing as the kilo lab is. Examples of suchapparatus are fluid bed crystallizers, hydroclones for theevaluation of that method of solid=liquid separation, lyophili-zation cabinets with special vial sampling capabilities, inter-mediate scale membrane processing assemblies, etc. An areawell suited for such testing purposes is not only highly desir-able, but often facilitates preparative work by processingmethods not within the scope of the kilo lab. Such an areashould be reasonably open for the manipulation of portableequipment, with ample walk-in hoods and tall Californiaracks, well distributed utilities, portable measurement panelsfor recorders, flowmeters and the like.

C. The Pilot Plant and Its Objectives

The objectives of the pilot plant environment in bulk drugprocess development are simple:

1. provide ready access to scaled-up processing for thebulk drug preparative effort;

2. give the preparative effort a responsive environmentto deal with the vicissitudes of the drug developmentprograms being supported;

3. permit the rapid and convenient evaluation of pro-cesses, methods and equipment, as well as the mas-tery of their scale-up;

4. obtain process data to support process design andthe development of procedures for the eventualmanufacture of the drug;

46 Rosas

Page 62: Active Pharmaceutical Ingredients

5. demonstrate process performance at a scale thatminimizes risk and the need to trigger scale-upconstraints upon first manufacture.

Of the above, only the last seems to require elaborationat this point, as process design is amply discussed in Chapter3. Thus, given a sufficiently large scale of processing in thepilot environment where the development effort takes place,the transfer to manufacture will be far less likely to entailscale-up risk and, more importantly, far less likely to createa regulatory scale-up issue upon first manufacture. Bulk drugpilot plants of recent construction at R&D drug companiesand at the major contract manufacturers provide batchprocessing vessels up to 10,000L for chemical work and upto 20,000L for fermentation work.

Table 3 outlines the equipment capabilities for a broadlycapable bulk drug pilot plant, such as can be found in themajor drug R&D companies, albeit not necessarily all in thesame location. Such plants have generally resulted by accre-tion and as needed to support vigorous drug development pro-grams; say, more than 10–15 compounds among preclinicaland all phases of clinical work. Indeed, pilot plant capabilitiesof similarly broad scope can also be found in the premier con-tract manufacturing companies, as they are active partici-pants in the preparative work for the whole spectrum ofdrug R&D companies.

Although in the 2000s the outsourcing field is populatedwith a greatmany small firms claiming to have cGMPpilotplant capabilities, Table 3 and the desired capabilitieslisted just below make it clear that the drug developmentbusiness is one in which size doesmatter. Obviously, thereis a sliding scale of capabilities vs. scope of physical plant,and Table 3 describes its full range so as to decisivelyaccomplish all of the above aspects of pilot plant work forbulk drugs. Lesser options entail lesser preparativepower, narrower range of scale-up and processing technol-ogies, and processes tailored to fit existing capabilities.Alternatively, a combination of outsourcing and in-houseresources can be used, but most often with far less agilityof preparation and development.

Process Development 47

Page 63: Active Pharmaceutical Ingredients

Physical plant by accretion means that, short of largelumps of capital investment, the processing capabilitieswill span designs and practices evolved over decades.However, issues of industrial hygiene and regulatoryexpectations have gradually done away with open proces-sing areas and rows of vessels in favor of processing mod-ules in various degrees of segregation and connectivities.Figure 14 describes one such prototype of modular designfor bulk drug chemical processing.

This table attempts to list most, if not all, of the equipmentfor a comprehensive bulk drug pilot plant. Accordingly, it is awish list requiring a great deal of capital investment for fulfill-ment. Nevertheless, such pilot plant ‘‘complexes’’ do exist, aris-ing mostly by accretion over decades, but also from projects oflarge scope used to augment and modernize earlier physicalplant obtained by accretion. Pilot plant investment is generallyviewed by R&Dmanagement as strategic, reflecting their long-term assessment of the vigor of the new drug pipeline, as wellas their unwillingness to accept preparative bottlenecks.

Some capabilities are needed very infrequently and arebest secured through vendors, other companies, or universitiesthat might possess them. For example: molecular distillation,high vacuum fractionation, gas=solid elutriation at scale, flui-dized bed crystallization, gas=solid catalytic reactors, vacuumbelt filters, gas=solid ball mill reactors (a la Kolbe), very highpressure stirred autoclaves and similar equipment. However,some projects reach the point at which the case for the in-housecapability becomes compelling, particularly if the technologyat hand seems attractive at large. Examples of such technolo-gies are: fluidized bed crystallization for the resolution of enan-tiomers, gas=solid catalytic reactors for selective oxidation ordehydrogenation of heterocycles, permeable wall tubular reac-tors and membrane-aided liquid=liquid extraction.

Similarly, hightly specialized capabilities and new tech-nologies are best provided at an intermediate bench=pilot scalefor ready evaluation of whatever advantage might drive thelarger scale proposition. This is a large part of the rationalefor the ‘‘bench=pilot’’ processing area, ninth in the list below.

48 Rosas

Page 64: Active Pharmaceutical Ingredients

For the smaller organizations, the appropriate pilot plantcapability presents a formidable challenge. Preparative powerand the ability to handle varied and often unpredictable pro-cessing tasks hinge on owning or having ready access to a suf-ficient breadth of processing equipment, preferably in acontext amenable to experimentation at scale. Absent thesecapabilities, the processes so developed are, inevitably, highlyconstrained in their scope and technical ambition; compro-mises are inevitable and timeliness and assurance of bulkdrug preparation trump any other consideration. As to out-sourcing as an adequate complement, its limitations are oftensevere, a principal topic discussed under VII.

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing

InfrastructureWarehouse space—partitioned and protected to meet cGMP and safetyrequirements

Materials handling suitable to the scale and scope of the processing tasksUtilities systems or suitable distribution from site systemsSteam (up to 11 atm)Cooling water (variable temperature depending on the source andseason)

Chilled water (down to 5�C)Refrigerated coolant (down to �25�C)Compressed air (up to 5 atm)Nitrogen (up to 2 atm)Portable hot fluid recirculating system (up to 250�C)Portable cryogenic recirculating system (down to �100�C)Electrical power (AC of normal voltage and of voltage required byindustrial motors; e.g., 110–440V in the United States)

Fire protection (no less than that required by the applicable codes)HVAC (rather variable according to space being ventilated)Tank farm (solvents, acids, bases)—all above ground, dikes as required

Pollution abatement systems (may vary widely in scope according to sitecircumstances)Acid=base neutralization capabilities (at source)Scrubbers (water and aqueous base) for the processing areasCarbon adsorption systems for specific emission pointsTrim condensers as required in processing ventsThermal oxidizer and stack for process vent emissionsTanks (segregated by waste category, dikes as required)

(Continued)

Process Development 49

Page 65: Active Pharmaceutical Ingredients

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

Ducting and fans for tie-ins to the various abatement systemsHazardous waste storage and unloading to haulers

Chemical processing areas: Nine distinct processing areas should beconsidered during the design or longer term planning of acomprehensive pilot plant for bulk drugs:

1. General chemical synthesis processing. Processing duties that do notfall squarely into any of the areas below

2. Finishing area for bulk drugs (includes solids processing). Lightchemical processing, if at all. Key duties of this area are to crys-tallize the final compound as the bulk drug (with the desiredchemical and physicochemical attributes). Includes filtration, drying,milling, classification, compacting, blending and packaging. Environment is distinctly cleaner than most other areas

3. Aseptic finishing area for bulk drugs (includes solids processing). Nochemical processing other than salt formation. Includes all of theabove finishing area provisions, but largely in an aseptic processingenvironment for the preparation of sterile bulk drugs. Requiressterilization equipment, special ventilation systems and much greaterpartitioning of the space

4. Hazardous processing for toxics, hydrogen, nitration, sulfonation, etc.Segregated operating space with extraordinary fire, explosion ventingand ventilation provisions. Contains the key equipment for reactionand limited work-up

5. Highly potent compounds processing. General processing equipment ina segregated area and equipped for a high degree of containment ofmaterials being handled, due to industrial hygiene and environ-mental safety reasons

6. Housekeeping (neutralization and other disposal activities with wastestreams). Complements the above areas, sometimes being within oradjacent (e.g., Fig. 14)

7. Fermentation processing. Very distinct in space, equipment andauxiliary facilities (16). Microbiology lab, seed development lab, andfermentor train. Air compression, air and liquids sterilization, tankand piping sterilization. Stirred tank fermentors, feed tanks andharvest tanks

8. Downstream processing of fermentation streams. Also a very distinctprocessing area: little chemistry but a great deal of work-up,purification and isolation with a different mix of unit operations (17)

9. Intermediate bench=pilot scale lab for engineering studies (not the kilolab, although it can be readily pressed into preparative duty asappropriate). Multilevel open bay space, walk-in hoods, tall racks,utilities stations for rented portable equipment, very little fixedequipment

(Continued)

50 Rosas

Page 66: Active Pharmaceutical Ingredients

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

Clearly, each of these areas has different requirements, but it is not inthe scope of this chapter to attempt a discussion beyond the aboveoutlines (23).

Processing equipmentGeneral purpose stirred vessels in the 100–5000L range. Glass

lined=316L stainless steel=specialty alloy in an approximate ratio of1=0.4=0.1 in frequency. Vessels above 100L should have split jackets. Allvessels intended for a processing function (as opposed to wasteneutralization, solution make-ups, etc.) should have variable speed drivesfor their centerline agitators and be baffled accordingly. Vessel layoutand connectivity can vary widely, but organization into multipurposeonce-through gravity modules seems to be the most useful for pilotingpurposes (i.e., from top to bottom levels: set-up, reaction, work-up,crystallization, and solids=liquid processing and housekeeping). Pleaserefer to Fig. 14

Within the category of general purpose vessels, a varietyexists that the design can put to good use. For example,vessels intended for crystallization will often haveagitator impellers better suited for that purpose, orvessels intended for work-up of reaction outputs willoften be fitted with auxiliary devices for liquid=liquidextraction or for evaporative concentration.

Fixed auxiliary equipment for general purpose stirred vessels (sizedaccordingly): condensers, decanters, receivers, weighing tanks, solidscharging devices, sampling devices, pumps and piping, connectivity to thetank farm and to pollution abatement equipment, connectivity amongeach other, vacuum sources, vent trim condensers, overhead catch tanks,in-line filters, flow splitters, etc.Portable auxiliary equipment for general purpose processing: pumps of

various kinds (centrifugal, positive displacement, vacuum), drumhandling devices with pumping provisions for charging to vessels, scalesof various ranges, stirred tanks (slant agitator) in the 100–1000L range,recirculating sampling or sensor loops, small filter press or pressure platefilters, blow charge tanks, line mixers, etc.Specific processing equipment and their auxiliary equipment (as above):

a. High-pressure reaction stirred vessels (glass lined up to 5 atm, 316Lstainless or specialty alloy to 15 atm).

b. Liquid=liquid extraction and phase separation devices: centrifugallyaided, including those capable to deal with suspended solids (i.e., forfermentation broths processing); mixer-settlers, membrane coalesc-

(Continued)

Process Development 51

Page 67: Active Pharmaceutical Ingredients

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

ing filters, rotating internal columns, etc.c. Gas=liquid contacting devices (other than pollution abatement

devices): Overhead venturi contactors, high turbulence contactors,packed or tray columns, wetted wall columns, etc.

d. Evaporation and distillation equipment: falling film, long tube, andwiped film evaporators with their condensing, receiving and vacuumsources; fractional distillation columns with their accessories,vapor=liquid disengagement inserts in selected vessels, etc.

e. Adsorptive processing equipment: columns and accessories for ionexchange, chromatography and other solid=liquid adsorption, high-performance liquid chromatography systems (columns, tankage,influent delivery devices, sensors and controls), molecular sievesolvent dryers, etc.

f. Solid=liquid separation devices: centrifuges (center-slung dig-out (upto 240 0) or bottom drop (up to 480 0), horizontal axis and side discharge),pressure and vacuum filters (stacked disk, plate and frame, agitatedfilter-dryers), cross-flow filtration, polishing filters, etc.

g. Solids drying equipment: tray dryers (vacuum and air), fluid beddryers, vacuum tumble dryers (with assorted internals), stirredfilter=dryers, countercurrent solids=gas dryers, spray dryers, lyophi-lization systems, etc.

h. Solids processing equipment: fluid bed and rotating shell processors,assorted grinders and mills, compactors and extruders, classifiersand blenders.

i. Solids=liquid processing equipment: homogenizers, colloid and ballmills, fluid bed and rotating shell processors, etc.

j. Membrane processing systems: cross-flow filters, ultrafiltration,nanofiltration, reverse osmosis, and pervaporation

k. Fermentors and auxiliary equipment: stirred tanks with specialcooling and steam sterilization provisions, designed for ease ofsterilization and maintenance of sterility, gas sparging, higherthan usual power inputs through agitation, air sterilizers, feedtanks of various sizes, liquid sterilizers (16)

l. Portable equipment cleaning modules, clean-in-place provisions inmany vessels, fixed cleaning stations

Process control capabilitiesThe bulk drug pilot plant must execute unusually varied processing with

the requisite degree of control over the process variables, as well as haveextraordinary means for data capture on-line (directly from sensors oranalyzers in the equipment) and off-line (from samples tested in thelaboratory and by derivation from raw data; e.g., the supersaturationprofile of a batch crystallization, the performance of a fermentation cell

(Continued)

52 Rosas

Page 68: Active Pharmaceutical Ingredients

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

mass from off-gas data analysis, the changes in the agitation require-ments through the course of a reaction or fermentation, the heat balanceacross a condenser, etc.). Indeed, pilot plant work provides the oppor-tunity to gather engineering data on scaled-up process performanceduring development, thus facilitating the better process design decisions.Often enough, the scale-up data drive the development of the process in adifferent direction as well.Accordingly, most vessels and other equipment are provided with the

appropriate sensors (temperature, pressure, level, pH, rotational speed,flow rate, weight, conductivity, etc.) and on-line analyzers as required.Some of the sensors may be used for local read-out (the value of theprocess variable may be read at the location of the equipment) and asinputs to a process control system elsewhere (a control room where,among other things, the inputs are converted by the controllers tooutputs to valves, switches and actuators in the field). The rest of thesensors may be used for local read-out and control (the control device isalso at the location of the equipment), but may also share the read-outwith the control room. The option of operating through local controlexclusively, while still available in principle, is rather unlikely to befound, as even operating environments of modest scope have mostlystrived for some degree of remote control.

A sensor for the process control loop consists of these principalcomponents:� A sensor for the process variable at the appropriate point in the

process stream and equipment, e.g., a thermocouple that generatesa voltage as a function of temperature. The voltage is received by atransmitter (usually located at the equipment) that converts thevoltage to a signal recognizable by the next device, e.g., a current inthe 4–10mA range according to a preset calibration of temperatureto voltage to current.

� A controller (or control device) that receives the input signal from thesensor and transmitter, and compares the value of the processvariable with a target value (the set point) and sends out an outputsignal to adjust the variable as needed.

� An actuator (a valve, a rotational speed drive, etc.) that, in response tothe output signal from the controller, seeks to adjust the processvariable. For example, the actuator may be a control valve thatallows more steam to pass through and thus increase thetemperature of the process materials in the equipment.

This said, remote process control can be variously implemented, takingadvantage of the many scopes of control systems that are available.Ambitiously, a pilot plant processing area will have most of its process and

(Continued)

Process Development 53

Page 69: Active Pharmaceutical Ingredients

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

infrastructure variables controlled remotely from a central location, usingdigital control devices connected to, with many governed by, a substantialcomputer system. The latter monitors selected process variables andcontrols many of those, relying on schemes that range from individualloops, for which the set point is entered at will, to complete schemes (andtheir algorithms) that sequence and control the processing events from amaster set of instructions (often referred by the unfortunate term ‘‘recipe’’)or maintain the readiness of the infrastructure (HVAC and utilities). Mostambitiously, as well as unwisely, the designers of the control system mayreach out and link the system that controls the processing events withextraneous systems, such as those that manage in-process and QC data orthose that manage materials inventories and procurement. Such excessiveconnectivity is not needed for process control or process data gathering, butgreatly increases the burden of the inevitable validation task. Whileperhaps of value in a large manufacturing environments, such linkages anddata sharing aid the pilot plant task marginally at the cost of greatervalidation and system maintenance efforts.Provisions for ascertaining the identity, lot number, and release status of

materials, such as bar coding and the like, should not be viewed as excessive,but should be well isolated from the direction and execution of process tasks.Indeed, the objective of theminimalist approach is to reduce to theminimumthose connectivities that are superfluous to the basic task of a pilot plant:simultaneously prepare material and develop the chemical process.Indeed, a very adequate and prudent approach is to delegate to local

microprocessors (or programmable logic controllers) the lesser controltasks, for which modern equipment comes with fully developed andvalidated process control packages; e.g., automatic centrifuges for filtra-tion, manipulation of heating and cooling fluids in vessel jacket services,HVAC management systems, etc. In this lean approach, and to the extentpossible, the supervisory process control system (at the top of thehierarchy) simply triggers the actions of subordinate systems and, duringthe period of action by the latter, it may monitor the appropriate processvariables but does not control them. At the processing level, thesubordinate systems do all the manipulations under the benevolent gazeof the top system and fade out of the scheme once the task is done; theydo not link with the supervisory system except to acknowledge theinstruction to start or to indicate its completion. Also in this approach,the supervisory system operates at arms length with any extraneoussystems involved with materials or laboratory data management. Theobjective, of course, is to severely limit the range of unintended conse-quences associated with large sets of computer code controlling multipletasks and manipulating large amounts of data. As the regulatoryexpectations on the integrity of control and other software-based systems

(Continued)

54 Rosas

Page 70: Active Pharmaceutical Ingredients

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

approach the fastidious, simplicity in the design of schemes for processcontrol and data management becomes compelling and, hopefully, therewill be a persuasive minimalist among those making such design choices.The qualifier ‘‘to the extent possible’’ recognizes the fact that certain

tasks are beyond the ability of the individual equipment control package,as well as the desirability of setting up at will specific control loops andtheir set points, the sequence of events, alarms, etc.Striving for such simplicity is not to be confused with the now ascendantSOP-based method of processing (standard operating procedure). In thelatter, the execution of the processing can approach a veritable daisychain of SOPs that minutely dice the overall task. In the extreme, the so-called manufacturing document (or whatever term might be used)becomes little more than a log, offering no perspective on the processing.Worst of all, operating personnel understand what they are doing dimlyat best, and mining the document for troubleshooting or assessmentinformation is tedious and often unproductive.Portable control modules may also be used; these consist of recorders and

controllers to create local loops with the appropriate sensors and valves oractuators. Specialized analyzers can be used on ad hoc or permanent basisfor on-line analysis of specific process variables, often through a sampling loop.A laboratory for in-process control testing must be equipped and staffed

well, and managed to be responsive and convenient to the pilot plantprocessing areas. In the pilot plant environment, there is a large datagathering component that often results in large loads of in-process testingrelative to manufacturing operations.Finally, and still in the process control subject, it is important to provide

the technical and supervisory staff with suitable and convenient officespace and related work areas. In all chemical processing, regardless ofthe control scheme being used, the eyeball contact with the processoperations is important. This is the case, most of all, for the bulk drugpilot plant, as a great fraction of the activities are being done for the firsttime and repetitive processing is so infrequent.

TraninngTraining facilities and unrelenting training are an indispensable part of a

bulk drug pilot plant operation, aimed at safe processing, reliablepreparation of bulk drug and effective use of the opportunity to scale upthe process and gather the desired know-how. Given the experimentalnature of the pilot environment, those that run it need to beextraordinarily sensitive to the process to be run. In that respect, SOPshave a rather limited value, as only the most elemental actions arerepetitive; today’s batch incorporates significant differences fromyesterday’s and will be different from tomorrow’s, even if performing the

(Continued)

Process Development 55

Page 71: Active Pharmaceutical Ingredients

Besides the physical plant there are, of course, other cap-abilities and attributes for a successful bulk drug pilot plantenvironment. To wit:

1. A skilled team of chemical engineers and chemists tooperate the facility, with the depth to operate as aprocess development cadre, capable of addressingthe full range of technical needs of scale-up and pro-cess design work as well as day-to-day operation.

2. Close collaboration and ready access to bench devel-opment chemists, microbiologists, and engineerswho have project (drug candidate) responsibilitiesin the pilot plant as well.

Table 3 Physical Plant of the Comprehensive Pilot Plant for BulkDrug Processing (Continued )

same basic process. Thus, training must be based on the fundamentals,which can be effectively presented to all involved.The increased use of SOPs, driven by a regulatory preference and the

seemingly paramount objective of consistency, can, in the extreme, dicethe operating instructions so minutely (approaching a daisy chain ofSOPs) so as make it very difficult for the operating personnel at variouslevels to fully appreciate the scope of the overall task and the linkagesbetween its different parts. This, coupled with the prevalent use of greatdetail in the operating instructions, has led to documents that areunwieldy, replete with discontinuities and very hard to use as trainingtools or for troubleshooting or retrospective data mining.While undoing this state of affairs may, alas, not be possible, it is indeed

possible, and most advisable, to preface the formal operating instructionsdocument with a brief outline of the process and its procedure, written inclear prose (not instructions in the imperative mood) and accompanied bya flow diagram of the procedure in the context of the designatedequipment, as well as including some brief discussion of the objectives ofthe work to follow. For example, a statement such as ‘‘set up RE-302 fordistillation under total reflux prior to the application of steam its jacket,’’an instruction that in today’s documents may take a number ofsubinstructions and no less than a page, conveys quite clearly theoperational intent and was, at one time, quite sufficient for a skilledchemical operator. A preview of such preface by all levels of operatingpersonnel has multiple advantages, including alleviating the insidiouseffect of the disrespect for the operating people implicit in the prevailingmode of operating instructions.

56 Rosas

Page 72: Active Pharmaceutical Ingredients

3. A skilled team of chemical operators that aretrained unceasingly in the fundamentals as wellas in all that is new by way of equipment, proce-dures, policies and applicable regulations.

4. A support laboratory for close and responsive in-process, troubleshooting, and QA=QC support tothe pilot plant operation.

5. Ready access to the analytical R&D function of thebulk process development area.

6. For fermentation processes, the appropriate labcapabilities as per 4. and 5. need to exist. These dif-fer markedly from those of chemical synthesis (16).

7. A skilled materials management function with theappropriate tools for materials tracking, documen-tation, and security.

8. A dedicated maintenance and minor installationteam with adequate workshop and stores.

9. A skilled clerical support function, well trained onthe regulatory obligations of the pilot plant opera-tion and the requisite tools.

Figure 14 The multipurpose, once-through gravity plant for bulkdrug processing.

Process Development 57

Page 73: Active Pharmaceutical Ingredients

10. Internal skills in the environmental engineeringfield and the applicable regulatory milieu for thepilot plant facility, as well as ready access to theappropriate site or corporate functions.

11. Internal skills in the operational safety and indus-trial hygiene fields, as well as ready access to theappropriate site or corporate functions.

12. Management systems and a managerial tone thatfoster, and insist on safe and responsible opera-tion, strict maintenance of the physical plant, con-tinuous training and strict regulatory complianceas called for by the developmental activity.

13. A management that foster and maintains a pilotplant organization as a vibrant, engaged andhighly skilled component of a broadly based processdevelopment function in the bulk drug business.An indispensable obligation of the pilot plant man-agement is to ensure that no one forgets, under thepressure of serious operational and regulatorydemands, that the pilot plant is an experimentalenvironment with a major responsibility in thecreation of the process body of knowledge.

Finally, and as indicated in Table 3, the pilot plant pro-cessing equipment needs to be set up and tailored to faciledata gathering, well beyond the usual process variablesmeasurements—extraordinary sampling ports and devices,nozzles, and flow loops set aside for the insertion of infre-quently used sensors, recirculating sample loop modules, flowloops, etc. Modern pilot plants are usually well provided withprocess control systems that monitor, control, and haveresponsive sequencing capabilities. Such systems are veryadvantageous in what is, after all, an experimental environ-ment. One needs to be alert, however, to the possibility ofmaking the process control system too rigid in its operatingprocedures, and thus discourage the enterprising experimen-talist or data gatherer. Finally, the operating style of such amodern facility should still encourage old-fashioned eyeballcontact with the process on the plant floor.

58 Rosas

Page 74: Active Pharmaceutical Ingredients

In summary, the bulk drug pilot plant is a critical massof skills and capabilities within the larger critical mass ofthe bulk process development function. Obviously, the sizeand scope of the organization matters a great deal—bulk drugpilot plants with capital replacement values of over a billiondollars and operating budgets well over US $100 million peryear exist. Nevertheless, and although difficult, pilot plantsof lesser scope and ambition can be created provided therequisite skills and management systems are assembledcohesively and maintained well.

D. New Processing Technologies

The process development environment is optimal for theevaluation of new technologies and methods for bulk drugprocessing, as all the necessary elements exist and are wellpoised for the acquisition of new experience:

a. the aggregate of discipline skills;b. the interdisciplinary critical mass;c. the experimental capabilities at bench, kilo lab and

pilot plant scales;d. the working interfaces with process design and

manufacturing.

Yet, there is an element of risk that, albeit of a differentcharacter, may be seen as comparable to that encounteredwhen evaluating new technologies inmanufacturing.Whereasthe latter is burdened with rigid regulatory constraints thatmay ultimately quarantine or preclude the sale of productmade under test conditions, the pilot plant is comparably con-strained; not because of the regulations, but because of therisk to the supply of material to the drug development pro-grams. This risk is, for all practical purposes, regarded justas large as, or larger than, lost manufacturing output, as thereis a potential impact on drug development timeline.

Alas, the seemingly obvious solution of evaluating newtechnologies on a parallel track does not work well enough.At some point, the new technology or method needs to bereduced to practice at scale and the perceived risk arises—compromising the yield or quality of material made under

Process Development 59

Page 75: Active Pharmaceutical Ingredients

the test conditions or usurping preparative capacity for non-preparative purposes. In a vigorous drug development pro-gram, that capacity (technical personnel as well asequipment) is usually fully allocated. Furthermore, R&Dmanagement at large has no sympathy for such distractions,a utilitarian outlook that could be well justified.

The less obvious solution, however, is to evaluate the newtechnology in stages, not unlike a newprocess variation arisingfrom the development work, such as a different starting mate-rial, solvent, or catalyst, an improved purification or a fasterandmore reliable dryingprocedure for thebulkdrug.These lat-ter changes are routinely introduced to the preparativework ina deliberatemanner, but with the relative procedural ease thatcharacterizes the pilot plant environment. R&D managementknows, perhaps deep in its subconscious, that the bulk processdevelopment function merges the preparative work withconstant scaling up of newmethods, and that a very large frac-tion of preparative work is also experimental. With discretionand with an extra measure of deliberation, new technologiescan be similarly evaluated at no greater risk.

In that regard, the more experimental aspects of the pilotplant that have been described under III.B are very wellsuited. In addition, getting the moral, as well as othersupport from the manufacturing organization adds to theimpetus and justification of the apparent distraction of bulkdevelopment resources. This is even more important to thetechnology stance of bulk drug manufacturing under the cur-rent and foreseeable regulatory environment that unwittinglydiscourages innovation into pharmaceutical manufacturing.

E. Beyond the Pilot Plant

As the consolidation stage comes to a close—slower pace of pre-parative work and having provided the body of knowledge con-tribution to the assembly of the dossier—the bulk developmentteam shifts its focus to the technology transfer to manufactur-ing. Although its participation in the preliminaries started,or should have started, at the early development stageand continued, increasingly, through the consolidation stage,

60 Rosas

Page 76: Active Pharmaceutical Ingredients

now the time has come to demonstrate the bulk drug processperformance in the first manufacturing plant or plants, the lat-ter in the event of multiple sites of first manufacture. In suchcases, the bulk process is generally operated in one plantthrough the bulk drug, but a slip stream of penultimatecompound (the final intermediate) or the final compound inunfinished form is shipped to another site for final processingto the bulk drug.

There is considerable material to cover on what happensbeyond the pilot plant, and such is the subject of Chapter 3.

IV. THE PHYSICOCHEMICAL ATTRIBUTES OFTHE BULK DRUG

As one of the three basic tasks of bulk drug process develop-ment, defining and achieving the physicochemical attributesof the bulk drug is pursued throughout the development cycle.Unavoidably, this effort trails that of the chemical or fermen-tation process, since its target comes from the dosage formdevelopment effort.

The difficulty of the dosage form task cannot be underes-timated. Its need for making judgments with partial dataactually exceeds that of the bulk development task, as thecrucial feedback on the bioavailability and stability of itsdevelopmental materials cannot be obtained rapidly, notunlike the feedback that the bulk development team needsas to the suitability of the its bulk drug for the dosage formpurposes. Figure 15 is an attempt to depict the scope of dosageform development.

Not to be neglected is the packaging development placeddirectly downstream from dosage form development.Sometimes complicated by the fact that the primarypackage (i.e., that in direct contact with the drugproduct) may also serve as a drug delivery device (e.g.,syringes, eye drop dispensers, intravenous bags) as wellas by the issues of interaction between the dosage formand the package’s material or the long-term stability ofthe drug product within the particular package chosen,the dosage form development function is additionally

Process Development 61

Page 77: Active Pharmaceutical Ingredients

buffeted by marketing issues that range from the serious(acceptability by the patient, for example) to the see-mingly frivolous (for example, the marketer’s insistenceon a distinct tablet shape that, although harder tomanufacture, lacks any apparent redeeming value).

Albeit hampered by a traditional disciplinary dividebetween pharmacy and the disciplines of bulk process develop-ment, the bulk=dosage development interaction needs to startearly and intensely. Not only is the task difficult for the reasonsjust stated but also there is considerable scope to getting to afirm definition of what is needed. Figure 16 lists the physico-chemical attributes of bulk drugs that must be controlled inthe bulk drug process, either directly, such as particle size, orindirectly, such as solid surface area or hygroscopicity.

Those attributes are, of course, set by the very lastprocessing steps of the bulk drugs:

a. the last synthesis step (or the last purification step ifa fermentation=extraction process);

Figure 15 The scope of the dosage form development task. Thenotation ‘‘Do loop’’ refers to the iterative process by analogy to theFortran language shorthand.

62 Rosas

Page 78: Active Pharmaceutical Ingredients

b. the subsequent isolation (usually by crystallization),filtration, and drying;

c. the final solids finishing (size reduction, classifica-tion, blending, and packaging).

Due to the significance of the physicochemical attributesof the bulk drug to its bulk and dosage form stabilities, as wellas to the dosage form performance (mostly bioavailability), ithas become increasingly frequent to add a recrystallizationafter the isolation of the final chemical compound and thusgenerate the bulk drug. Although such additional processingis expensive (its yield loss is incurred with the costliest com-pound), there are significant advantages to consider:

a. The final compound isolation is relieved form thedual burden of simultaneously achieving all thechemical purity attributes and all the physico-chemical attributes.

b. The discontinuity makes the final bulk drug lesssubject to upstream variations from the morecomplex synthesis=purification=isolation step.

c. The final recrystallization can provide an additionaldegree of purification that may be reserved as insur-ance or be part of achieving the final purity.

Figure 16 The physicochemical attributes of a bulk drug.

Process Development 63

Page 79: Active Pharmaceutical Ingredients

d. The final recrystallization can be developed with asharp focus on consistent attributes such aspolymorphic content, crystal habit, particle sizedistribution, surface area, and bulk density. Theseattributes also define hygroscopicity and are impor-tant factors on bulk and dosage form stability.

e. Regulators are very fond of such recrystallizations(for the above reasons), which can be used to morepersuasively present upstream process changes forapproval.

The addition of such final recrystallization is depicted inFig. 17. Given the fact that practically all bulk drugs arecrystalline for reasons of processing soundness, purity, stabi-lity, and consistent physicochemical attributes, obtainingregistration of a bulk drug in any other physical stateusually requires a compelling reason.

As practitioners discover (or should soon discover), crystal-lization skills are paramount among the skills set of thebulk process development (and manufacturing) function.

Figure 17 The final stages of bulk drug processing. A final crys-tallization is often inserted largely, if not strictly, for greater controlover the physicochemical attributes of the bulk drug.

64 Rosas

Page 80: Active Pharmaceutical Ingredients

Such skills should be nurtured, perhaps even lavished, aswell as complemented with a comparable physical chemis-try capability in the analytical R&D function.

This seems a good point at which define the analytical R&Dfunction as farmore than the guardian of quality during thepreparative work, or the highly skilled developer of assayand related methods, or the arbiter of regulatory issueswithin the bulk process development. For successful bulkdrug process development, the analytical R&D functionmust be an integral part of the process team: elucidatior,troubleshooter, contributor to the solution of process pro-blems, and intimate partner throughout. Any lesser invol-vement in the process task or a lesser aggregate of skillsis a strategic disadvantage in drug development.

Finally, the bulk process and dosage form functions needto collaborate earnestly at the earliest and, if needed, bebrought together under irresistible force to overcome the tra-ditional disciplinary gap. For example, the crucial decision onwhich bulk drug form is to go forward (the sodium salt? themaleate? the dihydrate?) is best made when the bulk processteam participates and is able to contribute its resourceful-ness, lest the dosage form team abandon the better bulk drugform because they do not know that its difficult attributes canbe managed or fully overcome upstream. It is there that thoseattributes are set through the actual process in the final reac-tion step and in the finishing steps that follow to afford thefinal bulk drug.

V. THE PROCESS BODY OF KNOWLEDGE

In developing the process for a bulk drug, the need to gatherand properly organize a process body of knowledge iscompelling:

1. The dossier requires that the process foundation—chemistry, engineering, scale-up, bulk drug chemical andphysicochemical attributes, environmental impact, processcontrols, preparative and developmental history, and bulk=dosage issues—be readily available and well organized forthe assembly of the various individual submissions. Increas-

Process Development 65

Page 81: Active Pharmaceutical Ingredients

ingly, these submissions need to provide a level of bulk drugprocess validation; namely, a persuasive case that, on thebasis of the actual performance during the preparative anddevelopmental effort, the process is capable of disciplinedmanufacture and thus result in bulk drug output that isconsistently safe and efficacious.

Additionally, the dossier should have the documentarybasis for being able to include in the submissions a similarlypersuasive case that the proposed manufacturing plans aregenerally sound. To wit, that the intended manufacturingmilieu will do justice to the process needs as identified duringits development. Such bridge documentation, while not reliev-ing the technology transfer team from the burdens of what-ever inspection the manufacturing plant may face, greatlyfacilitates the task of preparing for such inspection and fordealing with it if it occurs. Just as importantly, the saidbridge documentation also provides the regulatory reviewerwith sufficient background to answer a number of probablequestions and thus avoid their being asked during the reviewcycle.

It is rather easy for practitioners, when faced with theabove tasks, to get thoroughly lost in the detailed ‘‘howto’’ without fully grasping the scope and objectives ofthe dossier on the process, which have been definedabove. As the literature on ‘‘how to’’ grows and seminars,workshops, and guidance documents proliferate, thepractitioner should first seek a clear understanding ofwhat it is that the submission review and the plantinspection basically seek to accomplish. The above para-graphs are an attempt to clearly define just that, andadditional background material, still at the usefully gen-eral level, is suggested (18,19).

2. The technology transfer to manufacturing demandsthat the process be well documented. Most important, andas a coalescence of the process know-how, a comprehensiveprocess document, written for the specific purpose of impart-ing knowledge, is a requisite for the tasks of:

a. process design;

66 Rosas

Page 82: Active Pharmaceutical Ingredients

b. project engineering design and construction;c. procurement of materials;d. preparation of start-up plans and operating proce-

dures;e. transfer of the in-process and QC analytical methods;f. assessment of the process safety issues in the specific

context of the plant: operational safety, industrialhygiene, thermochemical, and environmental safety;

g. assembly (and timely approval) of environmentaland other regulatory permits;

h. definition of the process start-up targets of yield,capacity, waste loads, etc.;

i. dealing with assorted other matters, such as thosearising from the plant’s insurance, etc.

It is, of course, unacceptable to bind together all mannerof development reports and send them over with a covermemorandum (part of what is aptly known as ‘‘over the fence’’technology transfer). Attachments are very important, but awell-edited document that is rich in content and aimed atguiding the downstream practitioners is the indispensablefirst vehicle for the transfer of the know-how. Not even themost thorough collaboration between development and manu-facturing can completely remedy the lack of the above com-prehensive process document intended for impartingknowledge.

There is no attempt here to gloss over the extraordinaryeffort and discipline required to turn out such documen-tation on a timely basis. The consolidation stage isintense, the gentle slope of the know-how curve notwith-standing. Yet, the quality of the technology transfer—onboth the short and the long term—is very much enhancedif such a document is available soon enough.

Conversely, and as indicated in Figs. 11 and 13, the jointeffort with the operating organization (planning andprocess design) unavoidably overlaps the actual develop-ment of the process, sometimes to a great extent; e.g., ifan early decision is made to build a new plant or substan-tially alter an existing plant, the downstream work can-

Process Development 67

Page 83: Active Pharmaceutical Ingredients

not await a sufficient definition of the process, and infor-mation needs to flow as the process is developed. This is avery demanding task for all involved that benefits fromconsiderable practice, significant skills of process designon both sides and from a spirit of collaboration, prefer-ably steeped in previous joint successes. Unlike otherchemical processing activities, new bulk drugs areexceedingly driven by the ‘‘time to market’’ imperativeand organizations that can significantly overlap processdevelopment with manufacturing readiness work havea strategic advantage. However, having such skills andpractices do not relieve the development team from theduty of comprehensively documenting the process at theearliest reasonable time.

3. After successful technology transfer, which must, ofcourse, be well documented also, the original process body ofknowledge serves as the foundation for management of thechange control system, for training of new manufacturing per-sonnel and as the basis for sound process improvement work.Indeed, significant second-generation processes are most oftenbased on approaches suggested and partially elaboratedduring the original development.

4. Although the interaction with suppliers and contractmanufacturers will be discussed more amply under VII, it isoften that process information needs to be transmitted to out-siders, including prospective licensees of the drug candidate.Indeed, this happens most likely during the developmentalphase as help is sought in the preparation of intermediates,the bulk drug itself or in further development of the processor an alternative route for which the outside collaboratormay be better positioned.

It is in such instances that having a system for continu-ing process documentation pays off in the rapid satisfaction ofneeds that may arise unexpectedly. Ideally, the materialshould exist in organized form so as to permit knowledgeabletechnologists to assemble and edit a preliminary process doc-umentation package in a matter of a few days and a full pack-age in, say, 2 weeks. Of course, the transmittal of internaldocuments ‘‘as is’’ is fraught with the risk of undue disclosure

68 Rosas

Page 84: Active Pharmaceutical Ingredients

and it is best to transmit documents assembled and edited forthe specific purpose at hand, a task hardly feasible if thematerial does not exists or exists disjointed or incomplete.

The obvious need to avoid undue disclosure of internalissues and business methods, names, distribution listsand the like, as well as to avoid transmitting informationextraneous or strictly tangential to the technical matterat hand, must not be confused with undue reticence. Ifothers are expected to properly implement in-house pro-cess know-how or to use it as the basis for an activity tobe done on our behalf or as part of an agreement or license,the disclosure of the technical information must be no lessthan sufficient: what works and what does not work, thetechnical rationale for the prior decisions, our best under-standing of the process issues and sufficient detail of meth-ods, process design calculations, and data. In particular,data on thermochemical safety, industrial hygiene, andenvironmental profile need to be fully disclosed.

5. Developing the appropriate intellectual property is alsogreatly facilitated by the continuing process documentation sys-tem being advocated herein. Laboratory notebooks and pilotplant log or batch sheets, while useful for assigning dates ofreduction to practice, compositions of matter, procedural detailsand for the identification of inventors, are generally inadequatesources of cohesive process information and history.

6. Finally, there is the organizational objective of fos-tering a professional climate for the process technologists tothrive. The rigors and satisfactions of authorship of scientificand technical documents arising from one’s own work are notto be underestimated; they contribute greatly to the indivi-duals and to the organization as a whole, even if intendedfor internal publication only.

The ready access to powerful computers has createdan environment in which databases and templates orexcessively formatted documents are quite seductive asa seemingly easier substitute to a system based on docu-ments composed in clear, informative, and persuasiveprose. Thus, in such tempting systems, the processknow-how can be thinly dispersed over an alphabet soup

Process Development 69

Page 85: Active Pharmaceutical Ingredients

of spreadsheets and form-like documents that, inevitably,lack the full benefits of reflection and perspective from anauthor (or authors) with a process story to tell or a pointof view to present as to how to implement a process. Suchtemptation should be resisted, as extracting useful andapplicable process knowledge from the former environ-ment is not possible without a substantial effort of retro-spective composition that would have been better appliedto the creation of true process documents.

Scientists and engineers, usually handicapped as writersby the focus of their academic training and by misconcep-tion as to the scope of technical writing, are destined tofurther disadvantage if nudged by managerial conveni-ence or by conformity into documenting their work as iffilling blanks in a form, or seeing the process body ofknowledge as an array of suitably filled pigeonholes.

The effort in setting down and organizing the processbody of knowledge should not trail the acquisition of theraw inputs, as tardy heroic efforts to properly documentaccumulated knowledge are invariably not as good as the taskdeserves. Figure 18 outlines the body of knowledge task onthe applied effort vs. know-how plane.

Figure 18 The process body of knowledge in the know-how vs.applied effort plane.

70 Rosas

Page 86: Active Pharmaceutical Ingredients

Finally, Table 4 offers an annotated template that, iffollowed with sufficient discipline, carries out the variousmissions of the bulk drug process body of knowledge. Of par-ticular value are the milestone reports and the specificissues reports, as they permit achieving depth and focus,while nurturing vigorous authorship by the process technol-ogists. Such documents are invaluable as part of the compre-hensive process documentation, as well as excellent rawmaterial for the bridge documentation of the dossier. Theirelaboration into external publications is usually a much les-ser effort than starting from raw data and status reports.The following examples illustrate the proposed reports onmilestone events and specific processing issues. Note thatthe titles of these reports have been composed by this authoras fictitious from literature sources or approximate from hisown experience with actual bulk drug process projects:

Table 4 The Scope of the Process Body of Knowledge and ItsApplications

Process Development 71

Page 87: Active Pharmaceutical Ingredients

ICI 194008. The benzaldehyde imine route to the aminetosylate precursor. Bench development and readinessfor scale-up (5, page 22)

MK-787 via the ADC-6 chiral route. Results and experi-ence from the first large-scale pilot campaign (C. B.Rosas, personal communication, 2003).

Efrotomycin. Whole broth extraction in the mixer settlerand in the centrifugally aided extractor. Results andrecommendations for process design at the Stonewallplant (C. B. Rosas, personal communication, 2003).

MK-401. Early environmental assessment of alternativesfor the trichloro precursor (C. B. Rosas, personal commu-nication, 2003).

MK-421. Large-scale synthesis of AlaPro in a continuousflow system. Process design and results obtained at thelarge pilot scale (20).

Diazomethane. Pilot scale generation by continuous reac-tion and scale-up criteria for the commercial scale (21).

LY228729. Kornfeld ketone route as the selection forscaled-up development (22).

These two kinds of reports, when added to well-designedstatus reports that issue regularly and not too frequently, pro-vide the basis for a repository of a well-organized body ofknowledge that can be used for the various objectives pre-viously defined. Indeed, such reports are the core of the bodyof knowledge, as they gather, coalesce, and make cohesive forapplication the great deal of data and experience gatheredduring all aspects of process development.

Other aspects of the system in Table 4 are:

1. All the reports and documents listed originate in thebulk drug process development area, which embracesthe disciplines and function shown in Fig. 12, and ispart of R&D at large.

2. All process documents intended for the processdesign effort originate from the engineering disci-pline (chemical and biochemical), which embraces

72 Rosas

Page 88: Active Pharmaceutical Ingredients

the thermochemical and environmental safety func-tions within R&D. Material from the other disci-plines is attached as required.

3. All process documents intended for the dossierassembly are generated specifically for that purpose,usually through a CMC function (as per the chemis-try, manufacturing, and control component of theNDA). Such function is within the bulk drug processdevelopment area and not within regulatory affairs.This latter function should not use other processdocuments for its purpose of assembling the dossieror attempt to edit regular process documents onits own.

4. The biobatch, although an event taking place in thedosage form development area (and documentedaccordingly), will usually generate the need to docu-ment the process and related history of the bulkdrug inputs used.

5. The process document is generated upon completionof the technology transfer to first manufacture and iscoauthored jointly by the bulk drug developmentarea and the recipient manufacturing organizations.Chapter 3 discusses this and all other aspects of thetechnology transfer in some detail.

VI. PROCESSING RESPONSIBILITY IN BULKDRUG PROCESS DEVELOPMENT

All chemical processing, whether on a large or a small scale,whether for high value chemicals or commodities, or for bulkdrugs, textile polymers, petrochemicals or household products,carries a risk to those that work in the industry, to peoplearound themanufacturing sites and beyond, and to the environ-ment: locally, beyond the locality, and at large. Indeed, the riskcomes about from multiple directions:

1. The hazards created by the chemistry itself: (a)intended and unintended energy releases, and (b) the varioushazards of handling the materials involved. Bulk drugs often

Process Development 73

Page 89: Active Pharmaceutical Ingredients

present a peculiar hazard, i.e., the relatively high potency ofthe desired biological activities, as well as the collateral activ-ities of the intermediate compounds and, of course, of thedrugs themselves.

2. The specific manner in which the chemical processesis implemented at scale. Most risks in chemical processing area function of the process design, the equipment design, andthe operating procedures used to manufacture the products.In other words, the same inherent hazard can be implementedat various levels of risk depending on the specifics of imple-mentation, and often enough details matter.

Hazard—a source of danger, of possible injury or loss.Risk—The probability of suffering a given loss or injury

from a hazard.

3. The local context in which the manufacturing pro-cess is implemented. First, there are factors, such as theproximity to populated areas, the direct impact on sensitivereceiving bodies of water or other valuable habitats, or a lessapparent impact on remote parts of the environment atlarge. Then, as a lesser subset of those risks are the variousstatutory and regulatory constraints that create liabilitypotentials or that may impede timely manufacture if notproperly addressed.

It is one of the prime responsibilities of the bulk drug pro-cess development organization to seek processes of accep-table levels of risk in both the chemistry and itsengineering, and to participate in the process designand manufacturing plans to see to it that their implemen-tation risk is sufficiently low. Of all aspects of technologytransfer, none demands more in terms of the develop-ment team thrusting itself downstream and seeking theclosest collaboration with the manufacturing organiza-tion. Clearly, the greatest opportunity for success existsat the developmental stage of the R&D process work,when the process is conceived and developed; engineeringlow risk into the implementation of a hazardous processis always the second choice for the bulk drug processdevelopment team and the collaborating process design

74 Rosas

Page 90: Active Pharmaceutical Ingredients

function. For example, much safer process alternativesseemed to exist for the process that led to the 1974 cata-strophe in Bhopal, India (24); one called for a differentchemical route and the other for a different processdesign of the original chemistry.

Incidents such as Seveso, Italy, 1976 (24,25) and Bhopalillustrate the potential for catastrophic events from aber-rant chemical processing and design, sloppy operatingpractices and incomplete knowledge about probable unin-tended events and consequences.

Generally useful practices in this aspect of bulk drugprocess development are:

� Early assessment to guide the process conceptionand choices. This implies availability within the pro-cess development organization of, or facile access to,laboratory capabilities to evaluate thermochemicaland environmental hazards. The evaluation of indus-trial hygiene hazards is aimed at the protection of per-sonnel, and is facilitated by the availability or access toadequate toxicology resources such as those generallyavailable to a research drug firm. This industrialhygiene context, however, differs substantially fromthat of evaluating the risk to patients taking the drugs,and a different subset of skills and methods applies.

� Continuing assessment as the process develops,including a vigorous interaction with the processdesign function and the manufacturing organization.For example, issues such as the choice of manufactur-ing site, which influences the risk, cannot be settledby the process development team alone, nor can theybe properly settled without the hazards assessedduring development.

� Reasonably early decision on the in-house vs. outsidemanufacturing choices, as the latter requires technol-ogy transfer and due diligence work, as well as theinevitably longer cycle for reaching the necessarytechnical and business agreements (more on thisunder VII below).

Process Development 75

Page 91: Active Pharmaceutical Ingredients

A. Thermochemical Process Safety

Most chemical processing operations have energy exchangesbetween process streams and the surroundings; processstreams are heated or cooled for various purposes and suchexchanges need to be safe. Heated streams must not exceedlimits that generate undue pressures or undesired chemicalevents, whereas cooled streams must not freeze and interruptprocess flows, or hamper a desired chemical reaction andaccumulate unstable intermediates.

A distinction needs to be made between limits observed tomaintain process performance and limits observed toavoid a hazardous operating condition. The above para-graph refers, of course, to the latter limits, as depictedin Fig. 19 using the safe processing envelope concept.

The thermochemical safety of chemical processing dealswith the safe handling of the energy released from chemicalreactions and with the prevention of unwanted releases ofenergy. Chemical reactants may, when converted to products,result in the transformation of chemical energy into heat, andduring such exothermic reactions the heat release needs to besafely managed. In addition, chemical process streams mayreach abnormal conditions that cause unintended exothermicreactions, with the attendant formation of unintended bypro-ducts and release of energy.

Hence, the objectives of thermochemical process safety asa distinct principal component of processing safety at large:

� Identify all intended energy releases and determinetheir magnitude, rates, and byproduct releases, suchas gas evolution and their composition. These deter-minations need to be made over the appropriate rangeof process conditions.

� Identify unintended chemical events and energyreleases for reasonable hypothetical situations (e.g.,excess temperature by loss of coolant or runaway,excessive evaporation of solvent, interrupted reactioncycle, etc.) and assess their magnitude, probable rate,and consequences with respect to containment, gas

76 Rosas

Page 92: Active Pharmaceutical Ingredients

evolution and, when indicated, the composition andtoxicity of the components of a plausible release.

� Identify and quantify the hazards of handling theprocess streams and materials with respect to shocksensitivity, flammability, explosiveness in air mix-tures, dust=air explosiveness, etc.

� Seek process development solutions to avoid or reducehazards. For example, one might seek an alternativereactant, a reaction medium that permits a lowerreaction temperature or, in the ultimate, a differentsynthesis scheme for the conversions at hand.

� Provide process design solutions to those hazards thatcannot be reasonably developed out of the process,thus reducing their risk to levels appropriately lowfor the operating context. For example, a hazardous

Figure 19 Processing limits for performance and for safety. Pro-cessing limits define the perimeter of the operating envelope thatresults in the range of desired process performance, whereas thesafety limits define the safe processing envelope perimeter giventhe identified hazards that lie beyond. For example, a distillationis to be carried out at 90–100�C, whereas the high-temperatureinterlock that shuts off the steam is set at 125�C because a signifi-cant exotherm initiates at 160�C.

Process Development 77

Page 93: Active Pharmaceutical Ingredients

nitration reaction may be implemented in a reactorsystem that does not use aqueous coolants, or that isequipped with a suitable quenching vessel, or with asufficient containment system, or using a continuoustubular reactor with large cooling surfaces and hold-ing a small volume of reactive in-process materials.Similarly, a process or portions of a process with ahazard of explosion is preferably operated in a plantsite that is distant from populated areas (vs. an other-wise more suitable plant site not as distant frompopulated areas); or a process with an identifiedhazard of aquatic toxicity in its untreated waste wouldnot be operated in a plant site that normally dis-charges to an aquatic habitat. In both cases, one willtake preventive measures to reduce the risk, but a riskdifferential will exist between the two plant sites.

From the above, andmost importantly, the practice of ther-mochemical safety far transcends the evaluation or theassessment of hazards. It also demands that skillful solu-tions to the hazards be provided so as to eliminate themor reduce their risks as required. While the reader mayview this statement as redundant or exceedingly tutorial,the fact is that a functional discontinuity between theassessment of thermochemical process hazards and theimplementation of the process frequently exists, creatingan ever present pitfall for the unwary, the sloppy, the over-whelmed, and the unqualified, and even organizationswith the requisite critical mass of skills and well-documen-ted procedures need to be vigilant to the gap. As in mostother aspects of bulk drug process development, theutmost integration of process development and processdesign is the best approach to thermochemical processsafety, organizational divides notwithstanding.

Additionally, the above reflects the fact that the samehazard (e.g., a reaction mass that can decompose explo-sively upon total loss of solvent) poses different riskaccording to the context of implementation. Thus, forthe example just given of a major hazard, a process

78 Rosas

Page 94: Active Pharmaceutical Ingredients

design based on operator’s attentiveness and simple pro-cess controls would entail a greater risk than a designbased on interlocking and redundant measures to pre-vent total loss of solvent as well as operator’s attentive-ness. To wit, the operational risk arising from a processhazard is very much a function of the specifics of theoperational context.

There are, of course, many other aspects of processingsafety that are unrelated to, or overlap with, thermochemicalprocess safety. Among the overlapping, fire and explosionhazards due to flammables handling stand out, whereas theunrelated (e.g., falls, burns, asphyxiation in enclosed spaces,static electricity, etc.) are generally addressed through theaggregate of well-established measures of operational safety,facility design, insurance policy expectations and applicableindustrial or building codes.

It seems best, even for the introductory scope of thischapter, that before approaching a more specific discussionof the fundamentals and the practice of thermochemicalprocess safety, the presentation of a broad perspective beattempted. Hence Fig. 20, in which the field is viewed froma sufficiently high vantage and that the reader is urged toexamine in earnest before going further.

Three key points arise from Fig. 20:

� The chemistry defines the overall scope of the hazards:the energy release potential of the reactants and othermaterials used, that of the reaction and process streamsgenerated and the toxicity hazard that attends to all thecompounds involved, whether inputs, intended, or gen-erated by aberration. Accordingly, chemical acumen isutmost in the assessment and follow-up of the hazardsdefined by the structures at hand.

� Upon assessment, a broader set of skills is needed. Willthe hazards be avoided altogether by a change in thechemistry or will its risk be sufficiently reduced by aprocess solution? Either approach requires engineeringacumen to determine that a process solution is notadvisable or probable, or to devise a suitable alternative.

Process Development 79

Page 95: Active Pharmaceutical Ingredients

� Finally, the implementation of whatever process isarrived at through development, and its indispensableprocess design collaboration, must go through furtherengineering analysis, by which all the applicable consid-erations must be pursued to the requisite level of detail:from the sizing of vessel relief and area explosion ventingon the basis of thermochemical and related data to theevaluation of risk scenarios that will dictate the neces-sarymargins of safety relative to overlapping safety, sitespecifics (e.g., weather precedents, proximity to peopleor valuable environments), applicable regulations,insurance policy expectations and all the way up to theprobable perceptions in the neighboring communities.

Figure 20 Thermochemical process safety in bulk drug processdevelopment.

80 Rosas

Page 96: Active Pharmaceutical Ingredients

Prior to the catastrophes in Seveso in 1976 (24,25) and inBhopal in 1984 (24), these admonitions would have seemedunwarranted and even melodramatic, but not any more.Finally, seeking relief in the small scale of bulk drug chemicalprocessing does not help, as both instances of chemicalprocessing operations gone badly awry were of small scale.

1. Hazard Assessment and Methods inThermochemical Process Safety

Thermochemical hazards are numerous and richly variedin kind, each requiring more than passing considerationand, if appropriate, an assessment by engineering designcalculations, simulation, experimentation, or both. The taskcalls for experienced good judgment, as the possibilities aretoo numerous. For example, an organic synthesis of six dis-tinct steps, with up to, say, 10 distinct intermediate struc-tures generated, might also have a total of 40 differentmaterial inputs and process streams. Experimental assess-ment of each is a large burden that, invariably, can be greatlyreduced by the said experienced good judgment.

Herein there is not, of course, the aim to comprehensivelypresent this subject. Indeed, the literature is ample (not sur-prisingly, most was written after the 1976–1984 experiences),and the serious reader is earnestly referred to various refer-ences, preferably in the listed sequence (24,26–28). Clearly,this is not work for the dilettanti, but for professionals willingto invest in acquiring and applying focused know-how in amultidisciplinary environment– too much is at stake. Simi-larly, firms engaged in bulk drug processing cannot approachthe work in half measures, or contract it out indiscriminatelyor unaware of the pitfalls of doing so.

Let us discuss another perspective, this time from a closervantage; that of the thermochemical hazards assessment.Firstly, as indicated above, the structures at hand provide veryuseful leadsas towhat to expect.Asagoodruleof thumb,organiccompounds that are rich in nitrogen, oxygen, or both are high onthe list of reactivity and energy release structures, followed bysome specific bonds and then by the less obvious cases that exist

Process Development 81

Page 97: Active Pharmaceutical Ingredients

in organic synthesis, albeit less frequently (26, pp. 18–28; 27, pp.28–52;28,pp.22–27).Oncesoalerted, thehazardsassessorhasagoodnumber of techniques for estimation of heats of reaction, forrapid screening of exothermsand instabilities inmaterials, com-pounds and process streams, for accurate calorimetry workunder close to actual process conditions and for very specific fol-low-up of hazardous conditions (27, pp. 1–28 and 52–88; 28, pp.27–45). Indeed, the techniques are so numerous that care mustbe taken to walk the fine line between necessary and marginaltesting, striving to reserve the more elaborate and exhaustivemethods for the cases thatmerit them.For example, the thermalstability of process materials and streams can be pursued togreat lengths (29) as required. Similarly, the subsequenthazards of vapor or gas release or toxicity of the releasedmateri-als need to be pursued with similar acuity, as onemay go too faras easily asnot far enough. In this, the incisiveness of the screen-ing effort makes the difference.

The well-executed hazards assessment meets its threebasic objectives: (a) identifying and quantifying the heateffects of the intended chemistry, (b) identifying and quantify-ing, albeit not always as precisely, the thermochemical energyhazards from aberrant conditions, and (c) identifying andquantifying those hazards associated with the handling ofprocess materials and their instabilities. Again, it is workwith a great many nuances for which chemical, physicochem-ical, and engineering acumens are indispensable.

By way of vivid illustration of these assertions, one mightconsider the following instance, in which a labile nitrogen-rich compound was isolated as a water-moist powdery solidand dried under vacuum at�50�C. These latter drying con-ditions had been set at�50�Caway from the rapid and largeexothermic decomposition of the compound, found to initi-ate at�100�C in the screeningwork. Additionally, the heat-ing medium used in the drying step was limited to �55�Cand an ample vacuum capacity and a suitably low terminalpressure provided for the thorough removal of water. Aftermonths of processing at the ton scale, a process change wasintroduced in the isolation, substituting a mineral acid for

82 Rosas

Page 98: Active Pharmaceutical Ingredients

another in the final acidification prior to filtration andwashing. This seemingly innocuous change resulted in aproduct of slightly lesser purity that, alas, was significantlyless stable. The latter fact came forward upon violentdecomposition of �1 ton of product during the drying step.Subsequent investigation revealed that the process changeproduct was somewhat less crystalline and had a signifi-cantly earlier onset of decomposition, such that at 50–55�C the self-heating process of decomposition started andrapidly took the material to its violent outcome.

Finally, thermochemical hazards assessment needs tostart upon scale-up to the kilo lab, and if the structures athand are suspect, some basic screening should be done evensooner. The effort then needs to continue as the process isdeveloped and scaled up to the pilot plant, ensuring thatsignificant process changes are not missed—a task of skillfulvigilance, as the above example emphasizes.

2. Process Design from the Assessed Hazards andAchieving an Acceptable Risk

As indicated in Fig. 20, the hazards assessment data need tobe placed in a process design context, in which scale issuesarise forcibly: loss of surface to volume ratio, longer timecycles of certain batch events, more difficult mixing, largerin-process inventories, and many other. Upon scaled-up development, a reasonably specific design of the scaled-up operation needs to be challenged by the hazard and theresulting level of risk evaluated. This requires a sufficientengineering input and a deliberation commensurate with themagnitude of the hazard, and the exercise resembles the do-loops of computational code, with the effort resulting in a pro-cess design solution deemed to have an acceptable risk. Oftenenough this analysis leads to: (a) a significant change in thebasic process (the scaled-up risk demands a lesser hazard) or(b) to a highly engineered design (the hazard is accepted, butits scaled-up risk is also accepted). Examples of these out-comes are the switch to a different reaction to get to the samestructure or the use of a continuous reactor (or skipping theisolation of a dry unstable intermediate), respectively.

Process Development 83

Page 99: Active Pharmaceutical Ingredients

Beyond the above, more detailed methods of analysisexist for final plant design (e.g., HAZOPS or similar methods(30, pp. 42–178), with the objective of ferreting out the risksarising from the basic process hazards as well as all otheroverlapping hazards in the specific context for the processoperation. In many cases, the specific risk analysis resultsin changes to the safe processing envelope so as to deal effec-tively with the risks. This result is depicted in Fig. 21, wherea contoured envelope is adopted so as to place greater ‘‘dis-tance’’ between the permissible ranges of process variablesand the risks. For example, the mixed acid concentration ina hazardous nitration may be lowered to reduce significantlythe risk of catastrophic failure by corrosion of the preferred(and existing) reactor vessel in the plant.

Eventually, this continuing exercise embraces all theissues of the context of choice: the intended production capa-city, the intended operating space, the plant site locationand their myriad specifics. This is one of the principal rea-sons for having a close collaboration between the bulk drug

Figure 21 The contoured safe processing envelope. Detailed ana-lysis of hazards in a specific context (specific process and plantdesigns) usually reveals specific risks, and implementation of theprocess in that context may require that the original safe processingenvelope be contoured (i.e., modified for the said risks). In the fig-ure, the original envelope has been shaped so as to create a distancefrom the risks that bears proportion to the magnitude of the risk,which is depicted by the size of each circular symbol for the risk.

84 Rosas

Page 100: Active Pharmaceutical Ingredients

development function and the process design function. Thelatter will usually be the most direct conduit to other func-tions at play, such as production planning, operationalsafety and environmental compliance. This desirable colla-boration, however, does not imply that the bulk drug devel-opment team is a lesser participant in the process designeffort; far from it, in the optimal scenario, the bulk drugdevelopment function possesses (and nurtures) sufficientprocess design skills.

3. Thermochemical Process Safety in TechnologyTransfer

By the time that formal technology transfer takes place, thethermochemical process safety issues are largely settled in thefirst manufacturing context, as described in the preceding para-graph. All that remains is the confirmation of the process perfor-mance and a reassessment of the risk on the basis of actualoperating experience in the commercial plant. Of particularinterest is how well the thermochemical process safety mea-sures have been dovetailed with the full set of operational andenvironmental safety needs at the plant site, as this latter set,while preoccupied with a very broad range of hazards requiringvery detailed measures (e.g., the lighting of exit signs, paintedyellow strips, explosion proofing of electrical equipment, iner-tion of vessels with flammable materials, etc.), overlaps withthermochemical process safety (and depends on the hazardsassessment data) on issues such as relief venting for runawayreactions, the lesser risk location of hazardous processing,emergency planning for chemical releases, etc.

B. Industrial Hygiene

Examples of serious harm to workers from materials used andmade in the manufacturing workplace are well known: blacklung disease and asbestosis stand out by the number of peopleaffected and the severity of the results to long-term exposureto coal dust and asbestos fibers, respectively. Thus it is logicalthat, when dealing with bulk drugs (chemicals with potentand multiple biological activities), and as with thermochemi-

Process Development 85

Page 101: Active Pharmaceutical Ingredients

cal process safety, industrial hygiene (IH) issues arise early inthe bulk drug process development cycle.

Unlike thermochemical process safety, however, the bulk ofthe IH effort (its hazards assessment and risk analysis) fallselsewhere, as the toxicology and preventive measure skills arenot in the bulk drug process development function at all. Never-theless, the bulk drug development function does have someimportant roles to play in ensuring the IH safety of its processes.

One of these roles is to be very alert to what chemicals areused (and their specific physicochemical properties) and ensurethat the requisite toxicology screening is done on a timely basis,as well as tracking relevant process changes during develop-ment for their appropriate IH assessment. If anything, the bulkdrug development team is very well positioned to be sensitive tothe IH issues of new drug candidates given their knowledge oftheir biological activities and, at the very least, of their in vitropotencies. The interaction with the toxicology function also pro-vides an early exposure to the toxicological profile of the bulkdrug as it develops. Thus, if a compound’s intended use is basedon its cytotoxicity, that sets the stage for its handling even at thesmall bench scale. Indeed, there are toxicology screens thatapply to the IHmeasures, since the latter are concerned not onlywith the bulk drug proper but also with all materials handledduring the preparation or manufacture of the bulk drug. Inmany cases, intermediate compounds are found to have undesir-able markers in these screens; e.g., dermal or ocular irritants,assorted acute toxicities, potential teratogenicity or mutageni-city, etc. The inevitable focus on the toxicology of the bulk drugmust be balanced by ensuring the proper examination of theintermediates and to do so at an early stage. It is not possible,as an operating premise, to treat all compounds as worst cases;the appropriate data are needed for the protection of the person-nel during development and for setting up the proper engineer-ing measures and procedures for first manufacture.

The other role of the bulk drug team is, of course, to engi-neer major IH challenges out of the process, not unlike theelimination of thermochemical process hazards. For example,the process development effortmay seek the avoidance of isola-tion of specific intermediates or of their handling as dry solids.

86 Rosas

Page 102: Active Pharmaceutical Ingredients

One might seek their fine particle distributions enlarged and,with the bulk drug proper, crystallization technology may beused to avoid milling of solids. Additionally, technologies ofcontainment have been developed to deal with compounds ofhigh potency in their biological activities at any scale of proces-sing, and such technologies need to be practiced in the well-rounded bulk drug pilot plant, as indicated in Table 3.

Finally, access to sufficient IH skills is needed by the bulkdrug process development function. Many IH issues are not inthe scope of the toxicology function of drug development andrequire an additional set of skills, overlapping with opera-tional safety and occupational health regulations as well. Inone instance, a seemingly adequate containment of fumesand ventilation in an area where phosphorus pentachloridewas handled could not prevent a very mild baseline irritationof respiratory mucosa of workers such that, upon theirsubsequent handling of a penicillin derivative in the nextproduction campaign, severe allergic reactions developed.Nevertheless, the practitioner of bulk drug process develop-ment should be more than just aware of the IH issues andis hereby referred to some suitable introductory material(24, pp. 22–81).

C. Environmental Safety

Here we return to a forceful and decisive role of the bulk drugprocess development team. Just as the chemistry sets thescope of the thermochemical process hazards, so it does insetting the environmental profile of a bulk drug process: itsinherent benevolence (‘‘green’’ chemistry) in one extremeand its highly engineered implementation, made possible byintensive and extensive abatement and waste treatmentmeasures as the other extreme.

Early assessment of environmental profile (or potentialimpact) is the best tool to steer the chemistry along a greenerpath. While the actual data are much harder to obtain forsome key components of the profile (e.g., aquatic toxicity), sui-table screens exist (31, pp. 93–177; 32). These, coupled withpreliminary process design as to waste loads and some

Process Development 87

Page 103: Active Pharmaceutical Ingredients

assumptions as to manufacturing sites, make it possible tofeed back to the synthesis conception any of the following:

� The profile is such so as to merit the immediate searchfor alternatives for some specific aspects of the synth-esis. The synthesis team may find such urging verydisagreeable, but organic chemists have come a longway in accepting such judgments, even as early esti-mates. Of course, much depends on the level of skilland recognition of the assessors, which is one of thereasons for the early environmental assessment effortto be carried out by qualified people within the processdevelopment function, where they are generallyperceived as less bureaucratic and regulation driventhan comparably qualified people in a corporate ormanufacturing function.

� The profile is promising and some particular aspects needadjustmentorearlyenvironmentalengineeringattention.

Following the early assessment, the parallelwith the ther-mochemical process safety effort is quite close, except for thegreater difficulty and longer time cycle of some of the key datagathering.The issueof riskasa functionof context, site locationin particular, arises more sharply than with thermochemicalrisks due to the greater variety of downstream impact issuesand of how far downstream they might arise. Questions ofimpact of the eventual discharges on seemingly remote andvaluable habitats can arise, particularly with residual concen-trations ofhighlypotentdrugs (e.g.,mutagens, endocrinemodi-fiers). More recently, the issues of drugs in drinking watersources and theultimate fate of drugs excretedbypatientshaveentered the regulatory expectations.

The companion Chapter 3 will revisit the environmentalsafety topic in the process design and technology transfer con-text. However, it can be stated herein that the environmentalprofile of the bulk drug process has moved up in the prioritiesof R&D as the drug dossier needs to address various levels ofenvironmental safety assurances in individual regulatorysubmissions. The new drug application (NDA) in the UnitedStates, for example, requires an environmental impact state-

88 Rosas

Page 104: Active Pharmaceutical Ingredients

ment of a scope that cannot be dismissed. It no longer sufficesto provide statements of assurance as to compliance with allapplicable environmental regulations.

Finally, there often are overlapping jurisdictions bearingon the ability of getting first manufacture started on a timelybasis. All need to be satisfied that the intended manufactur-ing will not adversely affect the respective environments, justas communities and environmental advocacy organizationsmay need to be reassured. For all these and the abovereasons, the environmental profile of the bulk drug processhas risen in its importance, making it a good business choiceto have a competent and well-quipped environmental technol-ogy function within the bulk drug development function anda close collaboration with the complementary environmentalskills in the manufacturing and corporate organizations.

VII. OUTSOURCING IN BULK DRUG PROCESSDEVELOPMENT

The last decade has seen a drastic transformation of the bulkdrug manufacturing milieu, including the adoption by theresearch-based drug industry of a business model (perhapsapproached as a gospel in some cases) that greatly reducesthe role of bulk drug manufacturing in-house and increas-ingly places it with outside suppliers. The latter have prolifer-ated in the rush to capture a more profitable business thatfine and specialty chemicals, and many are fully engaged indrug intermediates and bulk drug manufacturing, to thepoint that by 2002–2004 overcapacity exists.

Inevitably, this has had a major impact on the bulk drugprocess development as well. Given the manufacturing driverfor this shift, the overall outsourcing topic will be discussedin the next chapter, including the said impact on the bulkprocess development.

VIII. IN CLOSING

Clearly, this chapter has described bulk drug process develop-ment as a complex, richly textured activity that is deeply

Process Development 89

Page 105: Active Pharmaceutical Ingredients

rooted in scientific and engineering skill. The discussion hasbeen largely based in the context of a large drug companywhere all the requisite skills reside, mostly in R&D, but com-plemented well by those of the downstream organizations.

The reader may well ask, particularly as the new drugbusiness faces increased pressures to do it faster and in a moreregulated environment, does the smaller organization havea chance to succeed? What if the seemingly indispensablecritical mass is not there and, instead, the task must be doneby dovetailing as best one can resources and functions frommultiple organizations? To the author the answer is clear.Bulk drug process development is a business where size mat-ters and matters greatly, and if success is measured by thetimely introduction of new drugs (not just one drug at a time)on a broad marketing base, then the smaller organizationslabor at a disadvantage and the virtual company struggleswith projects of any scope. Indeed, for bulk drug projects ofunusual technical difficulty, the smaller organization seemsfaced with insuperable odds. Yet, none of this denies theopportunity for the bulk drug process developer practitionerto excel and find professional fulfillment in any environmentwithout regard to size; all that is needed is the requisite skilland dedication to one’s work, as well as reaching out for thebest possible collaborations that might be available.

REFERENCES

1. Pisano GP, Wheelwright SC. The new logic of high-tech.Harvard Bus Rev 1995; 73:93–105.

2. Gadamasetti KG. Process chemistry in the pharmaceuticalindustry: an overview. In: Gadamasetti KG, ed. Process Chem-istry in the Pharmaceutical Industry. New York: MarcelDekker, 1999:3–17.

3. Repic O. Principles of Process Research and Chemical Develop-ment in the Pharmaceutical Industry. New York: JohnWiley &Sons, 1998.

4. Anderson NG. Practical Process Research and Development.San Diego, CA: Academic Press, 2000.

90 Rosas

Page 106: Active Pharmaceutical Ingredients

5. Lee S, Robinson G. Process Development—Fine Chemicals fromGrams to Kilograms. Oxford, UK: Oxford University Press, 1995.

6. Atherton JH, Carpenter KJ. Process Development—Physico-chemical Concepts. Oxford, UK: Oxford University Press, 1999.

7. Laird T, ed. Organic Process Research & Development. ISSN1083–6160. Columbus, OH: American Chemical Society, 1997.

8. Flickinger MC, Drew SW. Encyclopedia of Bioprocess Technol-ogy. New York: John Wiley & Sons, 1999.

9. Calam CT. Process Development in Antibiotic Fermentations.Cambridge, UK: Cambridge University Press, 1987.

10. Atkinson B, Matuvina F. Biochemical Engineering and Biotech-nology Handbook. 2nd ed. New York: Stockton Press, 1991.

11. Saunders J. Top Drugs—Top Synthetic Routes. Oxford, UK:Oxford University Press, 2000.

12. Corey EJ, Cheng X-M. The Logic of Chemical Synthesis. NewYork: John Wiley and Sons, 1998.

13. Shinkai I, et al. Tetrahedron Lett 1982; 23:4903–4910.

14. Lin JH, Ostovic D, Vacca JP. The story of Crixivan�, an HIVprotease inhibitor. In: Borchardt RT, et al, eds. In: Integrationof Pharmaceutical Discovery and Development—Case His-tories. New York: Plenum Press, 1998:233–255.

15. Trost BM. The atom economy: a search for synthetic efficiency.Science 1991; 254:1471–1473.

16. Soderberg AC. Fermentation design. In: Vogel HC, ed.Fermentation and Biochemical Engineering Handbook. ParkRidge, NJ: Noyes Publications, 1983.

17. Verrall M, ed. Downstream Processing of Natural Products. APractical Handbook. New York: Wiley, 1996.

18. Mathieu M. New Drug Development: A Regulatory View. 5thed. Waltham, MA: Parexel, 2000.

19. Berry IR, Harpaz D. Validation of Bulk PharmaceuticalChemicals. Buffalo Grove, IL: Interpharm Press, 1997.

20. Paul EL. Design of reaction systems for specialty organicchemicals. Chem Eng Sci 1998; 43:1773–1782.

Process Development 91

Page 107: Active Pharmaceutical Ingredients

21. Proctor LD, Wart AJ. Development of a continuous process forthe industrial generation of diazomethane. Org Process R&D2002; 6:884–892.

22. Martinelli MJ, Varie DL. Design and development of practicalsynthesis of LY228729. In: Gadamasetti KG, ed. ProcessChemistry in the Pharmaceutical Industry. New York: MarcelDekker, 1999:153–172.

23. McConville FX. The Real Pilot Plant Book. Worcester, MA:McConville, 2002.

24. Crowl DA, Louvar JF. Chemical Process Safety. Fundamentalswith Applications. Englewood Cliffs, NJ: PTR Prentice-Hall,1990:17–19.

25. Ramondetta M, Repossi A, eds. Seveso, 20 Years After. Milano,Italy: Fondazione Lombardia per l’Ambiente, 1998.

26. Stull DR. Fundamentals of fire and explosion. Am Inst ChemEng (AIChE) Monograph Ser 1977; 73:10.

27. Center for Chemical Process Safety. Guidelines for ReactivityEvaluation and Application to Process Design. New York:AIChE, 1995:9–173.

28. Barton J, Rogers R. Chemical Reaction Hazards—a Guide toSafety. 2nd ed. Rugby, UK: Institution of Chemical Engineers,1997:1–84.

29. Rowe SM. Thermal stability: a review of methods and interpre-tation of data. Org Process R&D 2002; 6:877–883.

30. Skelton B. Process Safety Analysis. An Introduction. Houston,TX: Gulf Publishing Company, 1997.

31. Allen DT, Shonnard DA. Green Engineering—Environmen-tally Conscious Design of Chemical Processes. Upper SaddleRiver, NJ: Prentice-Hall, 2002.

32. Venkataramani E, et al. Design of an expert system for earlyenvironmental assessment of manufacturing processes. Pro-ceedings of the 43rd Purdue Industrial Waste Conference.Boca Raton, FL: Lewis Publishers, 1989.

92 Rosas

Page 108: Active Pharmaceutical Ingredients

3

Bulk Drugs: Process Design,Technology Transfer, andFirst ManufactureCARLOS B. ROSAS

Rutgers University, New Brunswick, New Jersey, U.S.A.

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93II. The Process Design Task in Bulk Drugs . . . . . . . . . . . 96III. Technology Transfer of the Bulk Drug Process and First

Manufacture . . . . . . . . . . . . . . . . . . . . . . . . . . 106IV. In Closing—The Processing

Technologies of Bulk Drugs . . . . . . . . . . . . . . . . . 123Reference . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125

I. INTRODUCTION

This chapter complements its preceding companion chapter 2,which addressed the task of bulk drug process development.The tasks addressed herein overlap the development of theprocess, as process design does, or culminate the developmenttask, as technology transfer and first manufacture do. As inthe previous chapter, this one seeks to provide a soundperspective of the latter tasks to the uninitiated and the

93

Page 109: Active Pharmaceutical Ingredients

new practitioner, while the structured presentation and thedeliberately inserted points of view may interest and possiblychallenge the experienced practitioner.

First, there is the promotion of deliberately overlappingthe experimental development of the process with its designinto a manufacturing plant. Valuable as it is, however, thisoverlap is often not used as a powerful method in seekingthe better process and a manufacturing plant to match, butis practiced ineffectively, strictly as a necessity of the time-to-market imperative. Sometimes the jurisdictional divide atthe development=design boundary is too deep; or there is aninterdisciplinary gap, with chemists on one side and engi-neers on the other; or the process design becomes earnesttoo late to influence the development. Indeed, many scale-up difficulties cannot be identified or quantified soon enoughwithout a sufficient process design effort that runs paralleland close to the development.

Then, there is the lessened character that the processdesign subdiscipline has developed as the result of manybulk drug projects being handled by design and constructionfirms, where the practice of process design can be undulyconservative, or too pliant to the client’s wishes, or so lack-ing in the bulk drug processing skills so as to offer nothingbeyond what the client brings to the project, with the client’serrors or limitations dutifully incorporated into the design.In other projects, such as those that outsource manufactur-ing, the emphasis on process retrofit into existing plant isheavy and the process design, if any, is often beyond thegrasp of the client. This harsh assessment is warranted bythe penalties often paid, unknowingly at the time, becauseof the lack of the appropriate process design skills and prac-tices in scaling up bulk drug processes, or simply by theabsence of a mechanism to exploit the opportunities in delib-erately overlapping process development and design.

Alas, chemical process design skills are hardly evertaught formally; the first and last academic exposure mostengineering students have to the subject is a rather superfi-cial and highly structured ‘‘process design’’ project at theundergraduate level. To make matters worse, computer soft-

94 Rosas

Page 110: Active Pharmaceutical Ingredients

ware tools that can aid process design have usurped thatundergraduate task, often reducing the student’s effort tolittle more than filling blanks in fairly rigid templates, sometimes with proposed operational designs that can be hilarious(e.g., a stirred tank for a Kolbe reaction loaded with 4000 kgof 2-in steel balls!) and usually missing the learning experi-ence of manipulating design options at the conceptual level.Yet, sound process design is a requisite of good process per-formance in the manufacturing plant, and creative processdesign is practically indispensable in achieving superior pro-cesses and plants, as well as in exploiting advantageouschemistry that might be difficult to implement in the plant.Thus the wisdom of fostering the formal development ofthose skills and the development=design overlap inindustrial practice; placing emphasis on the conceptual andunstructured aspects, as these are not addressed well bythe current computational aids that are widely used, andare less likely to be pursued aggressively by engineeringdesign contractors.

Another objective of the chapter is to establish the valueof another overlap: process design is, for all practical pur-poses, the first stage of technology transfer. Moving a bulkdrug process from the development environment to that offirst manufacture is a delicate task that is made more difficultwithout a competent process design component. Once thesearguments are presented, the chapter is meant to flow rathernaturally as a series of annotated common sense prescriptionsfor sound technology transfer, proven to the author over num-erous projects and observations that ranged wide through thepractice of bulk drug chemical processing. We will dwell onthese measures as they apply through first manufacture,setting the stage for mature manufacture as a function ofproduct growth.

The chapter also includes, in closing, some observationson the technologies of bulk drug processing—development aswell as manufacture. While perhaps couched as pearls ofwisdom, they reflect some of the deeply held views of theauthor that could help the new practitioner with a perspec-tive of the bulk drug business enterprise.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 95

Page 111: Active Pharmaceutical Ingredients

II. THE PROCESS DESIGN TASK IN BULKDRUGS

A. Definition and Scope of Process Design

Process design is, first of all, not the development of the pre-cise specifications for a performing chemical plant, whetherbuilt on a green field or merely the modification of an existingfacility to accept a new process. Instead, process design takesplace well before such specifications can be drawn and it isonly through its completion that the plant design (in contrastto the process design) can be carried out, defining the futureplant to the extent that equipment can be procured andinstalled or an existing plant modified. Necessarily, processdesign has a much broader scope, including a largely concep-tual component that comes about early in the overall effort,confronting issues and unknowns in a sequence that is out-lined in the following sets of questions:

1. Broad brush definition of the task and its probablecapital cost, venues and timetable.

� At what approximate output will the process be firstrun and when? On what operating basis?

� What manufacturing cost can we project? What arethe top cost reduction targets?

� What are the probable materials and energy balancesof the process?

� Which operating site makes the most sense? In whatoperating area of the site of choice?

� Should it be run in a new plant or a retrofit?� What are the prospects for outsourcing some or all of

the manufacturing tasks? Which tasks are most likelyto be outsourced? When will that decision be made?

� What is the probable range of capital cost? Is the highend of the range acceptable? Is the low end (via out-sourcing) attractive enough?

� What is the probable timetable to an operationalplant? Is the timetable acceptable?

From dealing with these and other broad questions early,the process design should continue without pause as the

96 Rosas

Page 112: Active Pharmaceutical Ingredients

features of the process take shape with the benefit of feed-back, and the issues of site selection and outsourcing becomemore distinct. Indeed, as Fig. 1 illustrates, the fundamentalobjective of the parallel exercise with development is for thelatter to reflect the teachings of the process design.

2. Definition of the various process steps as they might beoperated in the most probable venue of choice. For example:

� Will the process run as currently operated at the pilotscale? At what scale? Batch-wise? Continuously?

� Integrated for optimal layout or placed opportunisti-cally throughout the existing plant?

Figure 1 Flow chart of the process design effort—bulk drugs: Theoverlap with the process development effort provides an unmatchedopportunity to seek the better process by using the feedback thatprocess design can provide. Both efforts move from the conceptualto the detailed and precise that is eventually needed to permit plantdesign, construction, and start-up. Similarly, as indicated by thehorizontal arrows between the two efforts, both feed forward andfeedback improve in defining the evolving process and its designas the efforts take place.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 97

Page 113: Active Pharmaceutical Ingredients

� With or without solvent recovery or recycle?� Are the environmental burdens acceptable?� Are there waste or pollution issues demanding

at-source treatment?� What are the identifiable risks from the know process

hazards? Where are the safe processing boundaries(i.e., the safe processing envelope described inFig. 19 of Chapter 2)?

� First flow sheets and their material and energy bal-ances are defined. What adjustments do they suggest?To the design or to the process?

3. More specific design issues arise. For example:

� How will these solids be separated from the highly vis-cous process stream? The pilot scale practice has beenan expedient not practical at the manufacturing scale.

� How will this large exothermic reaction be handledwithin the residence time constraints?

� How will this aberrant and unintended exotherm beprecluded? Or the associated risk reduced?

� The current method of isolation requires unprecedentedadsorption column diameters. Shall we seek alter-natives? Or should carry out more scale-up studies?

� Is this solvent throughput reasonable? Can concentra-tions be adjusted? Or should we use an internalrecycle loop via flash evaporation?

� Drying these particular solids seems intractable at theplant scale. Can we move the wet solids forward? Usea more volatile solvent wash to facilitate the drying?

� Availability of a vessel in the required material ofconstruction is a problem. Do we seek alternativematerials or alternative processing conditions?

� The industrial hygiene data for this intermediatedemands a given level of containment. Can the isola-tion be avoided?

Clearly, a myriad of such questions arise, preferablysooner rather than later. There will not be satisfactoryanswers to some, which should trigger subsequent iterations

98 Rosas

Page 114: Active Pharmaceutical Ingredients

in the feedback loop shown in Fig. 1. Although some extent offeedback from manufacturing planning always occurs, it isbest by far to establish the feedback loop as early as possibleand in a framework that makes the process developmenteffort sensitive to it. In other words, there must be responsive-ness within the development effort, as indicated in Fig. 1; par-ticularly when the issues that arise are inconvenient orundermine the more basic process choices that might havebeen made or that have the greatest appeal to the develop-ment team.

The best conditions exist where there is a process designcapability that collaborates with the process developmentteam; indeed, the latter should participate by virtue of havinga modicum of skills to understand the process design feed-back, as well as having the wisdom to act as needed. Conver-sely, the process design team must have sufficient knowledgeof chemical processing at large to understand the processimperatives or the rationale that makes a given processapproach so attractive so as to accept the design challenges.Frequent communication, even if at times burdensome, andoverlapping efforts are the key components of successful pro-cess design for a developing process. A new process, even ifpresented as developed, must survive the challenge of its pro-cess design; thus the compelling rationale for the overlap ofthe two tasks, as shown in Fig. 1. Another depiction of theresults of the overlap of process development and design isgiven in Fig. 2 for the example of a multistep process underdevelopment, showing the range of what happens as processdesign takes place: from straightforward implementation(Step 1) to iterative effort requiring change of the process con-cepts (Step 4) as indicated by the use of different letters andsuperscripts.

After considerable evolution, the principal finished pro-duct of process design is the process and instrumentation dia-gram (P&ID), eventually issued in what is usually called theapproved for construction version (AFC), the definitive suc-cessor of various intermediate versions and their revisions.A slice of such a P&ID is shown, with some simplifications,in Fig. 3. Obviously, there is a great deal of supporting detail

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 99

Page 115: Active Pharmaceutical Ingredients

that attaches to the P&ID AFC, but a critical examination ofthe diagram is the core of the subsequent plant design effort.

Perhaps the most difficult aspect of overlapping thedevelopment and design for a chemical process in flux is thatof the uncertainties from unsettled process issues, which mayrange, depending on the character of the process developmentorganization, up to the actual chemistry for part of the pro-cess. These uncertainties often appear in the P&ID as blankor ill-defined areas under the heading of ‘‘to be determined’’,as depicted in Fig. 4. Such uncertainties arise in the projectswith the most technical difficulty or in the fortunate instanceswhen a clearly superior and compelling part of the processexists but comes late to the fore, thus presenting the mostsevere test to the skill and discipline of the development=

Figure 2 Process design of a developing bulk drug process: Alongthe same lines of Fig. 1, process approaches (chemistry included),are indicated by the first letter in each box, versions within theapproach are indicated by the subsequent letters and elaborationsof a version by the superscripts, all used to depict the optimal evolu-tion of a process through the continuous feed forward and feedbackbetween development and process design. Note that Step 1 movedforward with little change, whereas in Step 4 a completely differentprocess approach was found necessary.

100 Rosas

Page 116: Active Pharmaceutical Ingredients

design interaction. Organizations or interacting teams thatcan manage those situations under the time-to-marketcompulsion have a major tactical advantage and, if they canexploit them under an enlightened R&D management, theiradvantage can be strategic.

Figure 3 The process and instrumentation diagram (P&ID):Shown as a simplified version stripped of some detail (to permitan uncluttered and legible diagram for reproduction herein), theP&ID identifies each item of processing equipment, their connectiv-ity and the control instrumentation loops. Not shown, but generallypresent in the final approved for construction version of the P&IDare the details of the piping, materials of construction, pump capa-cities, etc. Obviously, there is a wealth of other material that accom-panies the P&ID, but the latter is the centerpiece of the processdesign package used to execute the plant design.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 101

Page 117: Active Pharmaceutical Ingredients

There is, of course, the plant design effort, requiring alevel of detail that far exceeds that of process design and thatfollows it with considerable overlap, as depicted in Fig. 5. Weshould note herein that a bad process design cannot be turnedinto a good one by plant design means, as the latter are aimedat faithful implementation of the process design intent.

Finally, it should be understood that the discipline expli-cit in the above prescriptions does not depend all that muchon the size of the organization; it can be applied by the

Figure 4 The ‘‘to be determined’’ (TBD) provision in a P&ID:Although a very modest example is shown, the TBD provision is usedasneeded to indicateparts of theprocessdesign thatmaytrail theover-all design. This provision is particularly useful when the processdesignneeds tomove rapidly, sometimes at some risk that the outcomeof the TBD itemmay require redoing some of the related design work.

102 Rosas

Page 118: Active Pharmaceutical Ingredients

smallest team as long as the requisite complement of skillsexists within it or is sufficiently accessible and responsiveelsewhere. Indeed, one of the various arguments for havinga core of engineering skills within bulk drug chemical devel-opment is the ability to take snapshots of the developing pro-cess and do a good deal of informal and intimate processdesign or ‘‘back-of-an-envelope’’ design—e.g., is this the man-ufacturing plant we wish to operate? Or would this plant beamenable to ready expansion? Such snapshots permit swiftsifting of approaches and choices, more rapidly adjust thebench and pilot efforts, and spare formal process design effortfor more mature versions of the process. For projects withcapital or product cost sensitivities, the snapshots also permitrapid estimation of the alternatives.

Figure 5 Plant design as the sequel to process design: The abilityto compress the overall time cycle to first manufacture also dependson the extent to which process and plant design can overlap andpermit earliest plant construction activity. The instance of retrofitinto existing plant, although generally providing for the shortestcycle, can be adversely affected if the development and design over-laps miss an essential equipment item that is not at hand.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 103

Page 119: Active Pharmaceutical Ingredients

B. Process Design as the First Stageof Technology Transfer

Examination of Figs. 1 and 2 confirms the previous assertionthat process design is the first stage of technology transfer or,at the very least, provides the opportunity to initiate technol-ogy transfer to the advantage of the project. This is because inmost drug manufacturers the process design function (towhatever extent it applies to the project—new plant or retro-fit) is associated with the first manufacture of the product,and the know-how of the developing process begins to reachthe operational organization as process design begins. In bothfigures, the increasingly heavier arrows, as the project pro-gresses, indicates the know-how flow and the correspondingfeedback across the development=design boundary. This factis also apparent in Fig. 6, where process design begins whenthe process know-how begins to take its eventual shape.

Figure 6 An eagle’s eye view—from process development to plantstart-up: The earliest start of planning and process design formanufacture as overlapping activities with process developmentprovides the best chance of earliest first manufacture.

104 Rosas

Page 120: Active Pharmaceutical Ingredients

Participation in the development learning curve, reason-ably close for the process design function and somewhatdistant for the manufacturing function, can be very beneficialto the overall project, albeit at times the inevitable vicissi-tudes of process development cause distracting anxieties onthe downstream side of the development team—an occasionfor the appropriate managements to becalm the situation.Nevertheless, the opportunity for the operational side to pre-pare for the technology transfer and first manufacture isexcellent, and an exemplary mechanism for such transfer willbe presented in Section III below.

However, the current prevalence of outsourcing and thefrequent use of engineering design firms has created anenvironment in which technology transfer takes place in avariety of ways and some times not at all, as we will discuss.Alas, the tidy arrangement of doing everything in-house, asillustrated in the said figures, is gradually giving way to drugcompanies that manufacture only the very last stages of thechemical process. Nevertheless, the principles also illustratedin those figures are sound indeed, and good efforts to incor-porate as much of them into whatever development=designor development=first manufacture boundary applies areworthwhile.

C. The Process Design Demands on the ProcessBody of Knowledge

The demands of overlapping process design with the develop-ment of the chemical process are more immediate and some-what less rigorous than those of the dossier and of the finalprocess design or the formal technology transfer events.Instead, good communications are essential (again those backand forth arrows in Figs. 1 and 2), preferably complementedwith brief written reports when necessary. Apart from clarityand accuracy, timeliness is the next most precious quality ofthe information exchange that undergirds the overlap of pro-cess development and design. In other words, the snapshots ofthe developing process need to be rapidly examined throughprocess design as required by the scope of the changes beingintroduced or contemplated.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 105

Page 121: Active Pharmaceutical Ingredients

Beyond those demands, the process design function willeventually need the complete and fully organized body ofknowledge (Table 4 of Chapter 2) so as to permit:

a. process design;b. plant design (project engineering design and con-

struction);c. procurement of materials;d. preparation of start-up plans and operating proce-

dures;e. assessment of the process safety issues in the specific

context of the plant: operational safety, industrialhygiene, thermochemical, and environmental safety;

f. assembly (and timely approval) of environmentaland other regulatory permits;

g. definition of the process start-up targets of yield,capacity, waste loads, etc.;

h. dealing with assorted other matters, such as thosearising from the plant’s insurance, etc.

Aside from the procurement of laboratory equipmentthrough the capital project, the information needed for thetransfer of the in-process and QC analytical methods neednot go through process design and may pass directly to themanufacturing organization (QA=QC included).

Figure 7 depicts a proven model for the flow of the pro-cess body of knowledge as it is applied to process design andfirst manufacture.

III. TECHNOLOGY TRANSFER OF THE BULKDRUG PROCESS AND FIRSTMANUFACTURE

A. Definition and Scope of the TechnologyTransfer

Technology transfer has become the term that more appro-priately describes all the events associated with the firstmanufacture of a new bulk drug (or for that matter, of anynew product with its own distinct process for manufacture,

106 Rosas

Page 122: Active Pharmaceutical Ingredients

which will usually come from without the manufacturingenvironment). Older and not-so-old practitioners are probablymore comfortable with less comprehensive, but very descrip-tive terms, such as ‘‘process start-up’’ or ‘‘process demonstra-tion.’’

In its broadest definition, so as to capture all the activitiesfor its execution, the scope of bulk drug technology transfer

Figure 7 A model for the flow of the process body of knowledge:Successful first manufacture (timely, sufficient, and reliable)depends not only on the assembly of the requisite process body ofknowledge, but also on its timely flow to the various downstreamactivities. The model shown above has worked well in each ofnumerous instances that the author has seen it applied.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 107

Page 123: Active Pharmaceutical Ingredients

encompasses the tasks listed below. (Here the reader is encour-aged to place bookmarks on Figs. 11 and 13 of Chapter 2for perusal, as well as review the approximate Gantt chartfor the overall technology transfer in Fig. 8 below.)

Early stage (around the time the new drug candidateenters development):

(a) The operations area acknowledges the task of aprobable first manufacture tied to an expectation of regula-tory approvals to market a new drug.

(b) At the same time, the process development func-tion acknowledges its share of the above task—providingthe technology for the safe, dependable and timely executionof the first manufacture.

Figure 8 Gantt chart for the technology transfer of the chemicalprocess for a new bulk drug: The chart shows all of the key compo-nents of the overall task, with an approximate indication of theirrelative positions on the time-to-market cycle and, to a less preciseextent, of the relative widths of their timelines; the latter can varyconsiderably as a function of the process scope, new plant vs. retrofitinto existing plant and in-house vs. extent of outsourcing.

108 Rosas

Page 124: Active Pharmaceutical Ingredients

(c) The need for probable capital approvals and sub-sequent expenditures is forecast, presumably within the para-meters of an established longer-range plan that includes thelaunch of the new drug product as a probable event.

Next stage:(d) The process development function begins its colla-

boration with the operations area in addressing the broadbrush definitions of the project (as set out in II.A.1 above).

Next stage:(e) Overlapping process development, design and

manufacturing planning takes place.(f) Capital approvals are sought, in portions and from

a range forecasted for the total project.(g) Starting material sources are developed and busi-

ness terms negotiated (this may include extensive outsour-cing of, say, intermediates manufacture).

Next stage (some time after the biobatch milestone, butbefore filings from the dossier take place):

(h) The process body of knowledge is documented.(i) The final process design is completed.(j) The plant design is completed and installation

work proceeds.(k) Starting materials and auxiliaries are purchased.(l) Start-up plans and operating procedures are

developed.(m) In-process, QC, and regulatory methods (stabi-

lity) are transferred.(n) Process safety issues in the specific context of the

plant are settled: operational safety, industrial hygiene, ther-mochemical, and environmental safety.

(o) Environmental and other regulatory permits areassembled, filed, and approvals obtained.

(p) Definition of the process start-up targets of yield,product quality, capacity, waste loads, etc.

(q) The process validation plan is defined.(r) The process start-up team is assembled.(s) The plant installation is tested and readied for the

process.(t) The process is started up and validated.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 109

Page 125: Active Pharmaceutical Ingredients

(u) Preapproval inspections take place.(v) Process consolidation—the start-up continues to

demonstrate all targets under (p).(w) Results are documented, including updates oper-

ating procedures, in-process controls, etc. Heads of the start-up team sign off.

(x) Mechanical=instrumentation items punch-listand the ‘‘To Do’’ list are prepared.

(y) The start-up team is disbanded, but liaison per-sons are designated for matters arising. Manufacturing takesover.

The reader should beware that hidden within the abovereassuring list are all the necessary actions to solve unex-pected problems, particularly those arising during processstart-up, validation, and consolidation, or those in responseto significant observations from preapproval inspections. Dif-ficulties in technology transfer are inevitable; no such largenumber of activities that must dovetail precisely can go with-out some adversity or something being overlooked. Yet, well-executed projects for complex chemical processes generallymeet their targets of bulk drug deliveries and the existingprocess body of knowledge and assembled resources permitthe swift resolution of arising difficulties.

An approximate sequence of events following the desig-nated validation work (and its follow-up) that is useful forplanning purposes is (assumes a multistep process of signifi-cant scope):

During validation—25% of design capacity is reached.Month 2—60% of design capacity is reached.Month 3—80% of design capacity is reached. All lots are

without quality issues.Month 4—100% of design capacity is reached. All lots are

without quality issues.Month 5—procedures updated. Summary memorandum

on results is signed off and issued.Month 6—Operating personnel training confirmed (but

meant to continue). Comprehensive process start-updocument issued.

110 Rosas

Page 126: Active Pharmaceutical Ingredients

Obviously, the above timetable will vary with the scope ofthe process start-up: number of steps, number of plant sites,intrinsic process complexity, time cycle (e.g., long fermentationcycles plus downstreamprocessing plus any semisynthesis to fol-low). Sensible allowances to the above figures should be made.

B. Mechanisms for Technology Transfer

There are as many technology transfer mechanisms asthere are operational arrangements in bulk drug manu-facturing. However, the practice of technology transfer withinresearch-based drug companies can be said to take placewithin either of two environments:

1. R&D driven. In this arrangement, the R&D divisiondelivers, through its process development organiza-tion, a complete process to the operations division,which generally uses its process design function asthe principal gate to receive the process. While somespecifics may vary from instance to instance, thetechnology transfer (as just defined under III.A)takes place along the following lines:

R&D bears the principal responsibility for the technicalsuccess—it demonstrates its process to the operationsdivision. Accordingly, R&D leads the effort and casts aheavier vote on decisions bearing on the process and itsoperation, not unlike a first among equals. R&D also actsas the technical liaison with contract manufacturers andtransfers its process or chemistry to them, and eventuallysponsors the suitability of materials from those contractmanufacturers.

This environment offers the decisive advantage of a singlehandover—from the development activity in R&D to a per-forming plant that delivers bulk drug as required, and withreasonably well-defined responsibilities. Indeed, technologytransfer is an activity with all the vulnerabilities of a hand-over, and the analogy with certain sports is quite apt, thusthe advantage of a single transfer. Figure 9 attempts todescribe the R&D-driven environment for technology transfer.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 111

Page 127: Active Pharmaceutical Ingredients

2. Stage-wise. In this arrangement, the process develop-ment is split along disciplines or along operational lines, caus-ing more than one handover and with a greater spread of thetechnical responsibilities (Fig. 10):

a. The synthesis (or biosynthesis) and its analyticalcomponents are delivered by the R&D division (che-mists and microbiologists only) to an arm of the oper-ating division that has engineering and pilot plantresources. Obviously, the delivery of the chemistrymust be in stages as it develops, as the bulk drugsupply to drug development may be the responsibil-ity of the operations group. Eventually, the lattertransfers the process to manufacturing.

Figure 9 Technology transfer mechanisms—R&D driven: All ofthe process know-how flows from R&D organization to operations(manufacturing), outsources or both, although the diagram depictsthe in-house case.

112 Rosas

Page 128: Active Pharmaceutical Ingredients

b. The R&D division delivers the process to the opera-tions area at some intermediate stages ofdevelopment at which the chemistry has been estab-lished, with the rest of the development completed inthe operations area. The developmental bulk drugsupply is a shared responsibility, allocated accordingto ownership or control of pilot scale resources.

Regardless of the mechanism, however, all the activitiesunder III. A need to be carried out, even if less tidily. The sameis true for those frequent cases in which the sequence of

Figure 10 Technology transfer mechanisms—stage-wise acrossoperational boundaries: Either a partially developed process or adeveloped process (through the last intermediate material) is trans-ferred to the technical arm of the operations area, which carries outthe first manufacture activity, with R&D playing a secondary orcontingent role during the latter. Most often this mechanism resultsin two technology transfers.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 113

Page 129: Active Pharmaceutical Ingredients

process steps is divided among more than one manufacturingsite. Even less tidily, the same is true for projects with signifi-cant outsourcing, certainly to the extent that the processes arereliably established at each supplier.

In recent years, the advent of the USFDA preapprovalinspection method (and analogous inspections from agenciesoutside of the United States) have spawned the ‘‘launch plat-form plant’’— a multiproduct chemical plant designed for ver-satility, faster turn-around between products and capable ofmanufacturing a new bulk drug (or more than one new bulkdrug) until the full range of regulatory approvals and salesgrowth justify the transfer to another plant of larger capacityas a longer-term home for manufacture. This plant concept isdiscussed further in Fig. 11.

As to the technology transfer resources that must cometogether at the appropriate time, they range wide across bothR&D and operations:

From R&D—Process Chemistry and Process MicrobiologyChemical EngineeringAnalytical R&DCMC team that prepared the process input to

the dossier

Figure 11 The launch platform plant: A device aimed at avoidinga significant plant construction task by retrofit of the new bulk drugprocess into an existing plant dedicated to first manufacture only.

114 Rosas

Page 130: Active Pharmaceutical Ingredients

From Operations—Process DesignPlant design and installation (and

their contractors)Technical Services (divisional or from

the site of manufacturing)Production (from the manufacturing

site)Plant EngineeringQA=QCOperational, Health, and Environ-

mental SafetyMaterials Management (divisional

and site of manufacturing)

While some of the above players carry out crucial supportroles and are active in the day-to-day effort of starting-up anew process, the principal burden rests with those with theirhands on the actual operation, the process designers and theimmediate laboratory support (testing, ad hoc experiments toobtain a missing datum, validate a hypothesis on a problem,or to run a process manipulation in parallel to the plant).They have not only the task of demonstrating the processin new or modified equipment but also that of training theoperating personnel.

Some common sense and well-proven prescriptions are:

� A detailed log of events, preferably in clear Englishprose and with comprehensive entries, is essential.The ‘‘manufacturing operating instructions’’ orwhatever formal record of the processing is createdwill generally be far too structured as a series ofinstructions and blanks for data and signatures, dittofor logs from the control computer system, if any.

� As suggested above, the plant Technical Services labsshould be dedicated to support the process start-uparound the clock in speedy and unstructured waysthat QC cannot.

� The process start-up team should be well staffed innumbers and in the representation of all the skills

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 115

Page 131: Active Pharmaceutical Ingredients

and experience accumulated during the developmentand process design, set to apply more than sufficientpower to the task and make rescue missions unneces-sary.

� All background documentation, from the develop-ment, the process design and the installation shouldbe at hand and well organized for swift location ofneeded information.

� Operations management should keep its oversightdiscreet and be disciplined with respect to distractingthe start-up team for the latest, particularly at timesof stress.

� There should be at least one review meeting a day,attended by the principals of development, processdesign, analytical, technical services, production andproject engineering (installation), run sharply and tothe point, with ‘‘who does what by when’’ unambigu-ously defined.

� Data and other trends should be followed, preferablyfrom some premeditated plan, so that the directionof process performance can be assessed soonest.

Finally, a technology transfer device that works verywell in avoiding pitched battles upon process start-upproblems—where are the funds to fix the problems?—is tohave a fixed amount of the capital budget for the project allo-cated as a contingency to such fixes and under the control ofthe start-up team. This protects the process start-up from ashortfall of funds due to underestimation of the originalinstalled cost of the plant. A total of 5% of the total capitalbudget is usually a sound allocation for such contingency.The latter is, of course, apart from the usual 15% contingencyof such capital projects, and is given back to the corporation ifnot used (by keeping it out of the grasp of the plant manager).

C. Technology Transfer in the Outsourcing andLicensing Environment

Up to this point, we have alluded to the outsourcing environ-ment while describing systems and prescribing ways of

116 Rosas

Page 132: Active Pharmaceutical Ingredients

operating largely in the context of a big pharma organization,where all the capabilities exist under, ultimately, a singleman-agement. This has been useful in that it has permitted present-ing the tasks of process development and technology transfercomprehensively. Indeed, the tasks at hand are basically thesame and need to be done just as well without regard to howthe tasks might be divided between the various parties in thecurrent outsourcing environment for bulk drugs manufacture.

We need, however, to understand the complexities addedto the basic tasks by their becoming divided among:

� customers, ranging from big pharma to virtual and‘‘almost virtual’’ drug companies;

� suppliers, ranging from large fine chemical manufac-turers to small new companies with a claim to someniche processing technology;

� service providers offering to take only the customer’scompound structure and do it all through first manu-facture (generally relatively new and small compa-nies), thus appealing most directly to the virtual andalmost virtual customers.

Virtual drug companies are those that generally possessnothing more than the patents or the licenses to a com-pound, and operate by contracting all subsequent tasks.Almost virtual companies are those that, although havingdiscovery and some clinical development capabilities, lackeverything else.Starting with the big pharma customer, the technology

transfer task takes place in a relatively narrow range definedby the following poles:

� The customer wishes to outsource part or all of the bulkdrug manufacturing, using one or more contract manu-facturers. The customer also brings sufficient processknow-how and assumes the responsibility of demon-strating its performance in the manufacturer’s plant,according to a well-defined process start-up plan jointlydeveloped by the two parties (1). Figure 12 describes thishappy set of circumstances, clearly the best scenario for

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 117

Page 133: Active Pharmaceutical Ingredients

technology transfer between the customer (who has thetechnology) and the contractor (who as the plant and theright set of manufacturing conditions).

� The customer and the contractor come together on thepresumption that the contractor’s processing skills,plant capacity, and existing chemistry operations(with its available intermediates) constitute a signifi-cant advantage (time and cost) as a supplier of a givencompound, generally an advanced intermediate. Thecustomer may contribute all or part of the processfor the conversion of the contractor’s intermediate tothe customer’s target, although often enough the con-tractor does contribute the actual process.

Both poles define a range in which technology transfer isgreatly facilitated by the existence of a sufficiently devel-oped process from the customer (in the first case, Fig. 12)or, in the second case, by the relative ease in reachingthe target structure from the contractor’s existing inter-mediate, with or without some process development col-laboration.

However, on the other end of the spectrum, the virtualand almost virtual customers have no process technology oftheir own and thus seek one or more contractors that will

Figure 12 Technology transfer in the outsourcing environment—the optimal scenario, shown on the basis of the well-managed trans-fer of sufficient process known to a capable recipient.

118 Rosas

Page 134: Active Pharmaceutical Ingredients

make the compound on the basis of their existing offerings ofproximate intermediates. Here the customer has notechnology to transfer and the matter resides entirely withthe contractors. There is, however, a scenario that is increas-ingly found in the virtual and almost virtual customer domainand that can be best described as the ‘‘technology transferfrom hell,’’ which a mere examination of Fig. 13 will confirm.

Most cases of a compound being licensed to a virtual oralmost virtual company bring no process technology(beyond medicinal chemistry and preliminary prepara-tions) or partially developed processes. Thus, the licensoris, at best, in a weak position to transfer useful or completeprocess technology and usually lacksmotivation beyond theprecise letter of the license. Also in most such cases, thevirtual or almost virtual customer lacks processingbackground or experience in the fine chemicals milieu, orhires people with such competency much too late.

Figure 13 Technology transfer in the outsourcing environment—the worst scenario, shown on the basis of a virtual company thatlicenses a compound that does not bring a sufficient process bodyof knowledge.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 119

Page 135: Active Pharmaceutical Ingredients

While some of the above difficulties are inherent to thevirtual character of the customer, the latter can take someactions to avoid the ‘‘technology transfer from hell.’’ Namely:

� Hire or engage competent people in chemical manu-facture, preferably with experience in dealing withfine chemicals manufacturers, and preferably rightat the outset of mounting a serious clinical effort.

� Seek license language that unambiguously obligatesthe licensor to provide full documentation of the pro-cess and its experience with it (in a manner suitablefor input to a dossier), as well as an iron-clad obliga-tion to a serious technology transfer effort. Use mile-stone payments as a means to motivate the licensoron the latter.

The licensee should beware of accepting the licensor’s‘‘production documents’’ as the core of the processdocumentation, as such documents—operating proce-dures with the blanks filled in—are poor vehicles forimparting process knowledge. To the extent that itexists, the process body of knowledge should be welldocumented and provided by the licensor, along thelines described in Section V of Chapter 2.

� Obtain an independent evaluation (not from thelicensor or the potential contract manufacturers) ofthe development status of the bulk drug process rela-tive to its first manufacture and of the readiness ofthe resulting bulk drug for successful manufactureof the desired dosage forms. If development is notcomplete, seek its completion in a competent environ-ment rather than patching it up at the contract man-ufacturer or by hiring a modicum of staff to rush itthrough in hastily arranged laboratories.

� Manage its QA and regulatory team as earnestparticipants in establishing the bulk drug activityrather than approaching it strictly as a policingtask.

120 Rosas

Page 136: Active Pharmaceutical Ingredients

D. Regulatory Aspects of Technology Transfer

The principal regulatory task in the technology transfer of abulk drug process is its reduction to practice in accord withthe process defined in the dossier, and to develop and executea sufficient (not an excessive) validation plan. It is alsoimportant to do so at a contractor or contractors that under-stand the basis and procedures of the drug master file systemand their obligations to the customer’s dossier; having a GMPstatus is not sufficient, as the latter is the minimum require-ment. The customer should scrutinize those aspects of thecontractor’s operations early in the due diligence process,including its change control procedures.

Prepared for preapproval inspections (PAI) is not atrivial matter, as significant observations may delay theapproval of the corresponding applications; usually the PAItakes place after all other aspects of the application have beenreviewed and found suitable for approval. Although GMPissues may arise, the principal objective of the PAI is to deter-mine the soundness of the manufacturing process relative tothe process described in the application. Some golden rulesthat can do no harm are:

� Be prepared to credibly answer questions on the spot(all knowledgeable personnel immediately available,all documents organized and handy). Questionsshould not linger unanswered because of not beingprepared.

� Assume the inspector wants to know if you know whatyou are doing and that the scientific and technicalbackground of the process has been mastered by thosewho will run it.

E. Transition to Mature Manufacture

Successful transition to mature manufacture (different fromstatic or declining manufacture!) requires, of course, the firmbasis of a sound and well-documented technology transfer,including a list of all the ‘‘To Do’’ items (those actions of mod-est scope that would consolidate and improve the reliability of

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 121

Page 137: Active Pharmaceutical Ingredients

the process and its operation). The process training of theoperating personnel also needs to be consolidated, avoidingthe feeling of security suggested by their familiarity withthe production documents and the manipulations fresh offthe process start-up. On other words, consolidation of thenew manufacturing operation is the foremost objective aftertechnology transfer.

Next, the to do list should be executed promptly withinthe change control procedure. Measures for increasingproduction output should be conceived and taken as far asplanning in the event of product growth. If the latter isalready anticipated with some precision, those measuresshould be pursued deliberately.

If expedients with respect to raw materials were used toget to first manufacture (e.g., a single supplier of a criticalmaterial, a risky inventory position on another, etc.), thoseneed to be addressed immediately, particularly if expandedoutput is desired. On the other hand, superfluous requeststo qualify new suppliers (usually coming from Materials Man-agement) should be rejected for the time being.

Process changes for cost reduction should be pursued onthe basis of their technical soundness and merit first, then onthe basis of their cost impact, but mindful of the possibleintroduction of too many changes too soon, as well as of thevicissitudes of supplemental submissions to the regulatoryagencies. On the other hand, process changes aimed atincreasing output, while scrutinized just as much, could beput on a track faster than those for cost reduction if firm pro-duction targets for the plant justify it.

In evaluating the above process changes, the originalprocess body of knowledge should be mined with intensity,as invariably some good ideas and partially developedimprovements have to be set aside during the original devel-opment if the dossier target dates are to be met.

For intermediates or bulk drugs made by contractors, thesame approach embodied by the above recommendationsshould be sought in their operations, with particular atten-tion to their observance of sound change control proceduresand drug master file maintenance.

122 Rosas

Page 138: Active Pharmaceutical Ingredients

Figure 14 attempts to depict the sequence towardsmature manufacture just outlined in order of priority.

IV. IN CLOSING—THE PROCESSINGTECHNOLOGIES OF BULK DRUGS

It is not in the scope of this chapter to address this topic inany breadth and least of all in any depth. The variety of thetechnologies is too large and the field far too rich. Instead,some selected observations that the new practitioner mightfind useful follow.

1. Process development organizations that lack a suffi-cient engineering component often miss the opportu-

Figure 14 From first manufacture to mature manufacture: Theprocess change mechanism that leads, over time and various pro-cess and operational changes, to a mature manufacturing operationthat supports product growth and reduces the cost of goods.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 123

Page 139: Active Pharmaceutical Ingredients

nity of the better implementation of the chemistry byshunning continuous processing. It is not a matter ofdisdain, but of not having the tools and, oftenenough, the pull of the familiar batch or semibatchmethods is too powerful.

2. Product purity and consistency, which are para-mount norms in bulk drug manufacturing, aretoday observed through the impurity profiles to anunprecedented extent. This puts a great deal of pres-sure on the mastery of purification methods, mostlyon those based on crystallization from solution.

3. A final recrystallization of the bulk drug for thepurpose of a consistent composition of matter fromwhich the material emerges has considerable advan-tages in providing consistency of the physicochem-ical attributes of the bulk drug, including thecontrol of phase purity (single and consistent poly-morph). It also buffers the bulk drug from vagariesupstream and tends to becalm regulatory disquiet,particularly about process changes upstream.

4. Scale-up of chemical processes is a business of muchskill, largely because of the frequent intrusion ofphysical effects on the chemical kinetics. Good pre-dictive tools and solutions exist, however, to dealwith those intrusions by changing the physicalenvironment away from that of the bench scaleexperience, but requiring the application of chemicalengineering skills and the willingness to abandonthe familiar batch or semibatch stirred tank whennecessary or advantageous.

5. The bulk drug=dosage form boundary of processdevelopment is very difficult, often because of thediscipline differences and just as often because thedefinitive decisions on the dosage form side comelate in the cycle (often for good and largely unavoid-able reasons). The bulk team must be sensitive andskilled in delivering to the dosage team what theyneed, and get very involved with their issues early

124 Rosas

Page 140: Active Pharmaceutical Ingredients

in the development cycle, particularly to seek multi-disciplinary decisions.

6. Practically all prescriptions for sound and successfulbulk drug manufacture given in this chapter applyto the varied, seemingly tumultuous outsourcingenvironment, but only if a diligent effort goes intooperating, maintaining, and building trust in thecustomer=supplier relationships.

In closing, few industrial endeavors offer as many oppor-tunities for exciting and valuable technical work as the devel-opment of processes for bulk drugs and their implementationin performing chemical plants. The merging of chemistry (inits various fields), microbiology, chemical engineering, andpharmaceutics makes it possible, but demands that the prac-titioner of a discipline be earnest in the interaction with theothers, regardless of their disciplines or their functions. Onlythrough such effective interactions can success be reached inthe exciting and difficult business of bulk drugs.

REFERENCE

1. Pollak P. From a commodities business to the world’s leadingmanufacturer of exclusive fine chemicals. Chim Oggi 1997; 15:75–81.

Bulk Drugs: Process Design, Technology Transfer, and First Manufacture 125

Page 141: Active Pharmaceutical Ingredients
Page 142: Active Pharmaceutical Ingredients

4

Design and Construction of FacilitiesSTEVEN MONGIARDO and EUGENE BOBROW

Merck & Co., Inc., Whitehouse Station, New Jersey, U.S.A.

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . 127II. Business Requirements . . . . . . . . . . . . . . . . . . 129III. Developing the Preliminary Scope . . . . . . . . . . . . 131IV. Utilities and Building Systems . . . . . . . . . . . . . . 137V. Preliminary Scope Deliverables . . . . . . . . . . . . . 138VI. Design Development . . . . . . . . . . . . . . . . . . . 142VII. Utilities . . . . . . . . . . . . . . . . . . . . . . . . . . . 147VIII. Safety . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150IX. Current Good Manufacturing Practices Requirements . 150X. Qualification Plan . . . . . . . . . . . . . . . . . . . . . 151XI. Expansion Capabilities . . . . . . . . . . . . . . . . . . . 152XII. Hazard and Operability Analysis . . . . . . . . . . . . . 152XIII. Execution Strategy and Planning . . . . . . . . . . . . . 154XIV. Procurement Strategy . . . . . . . . . . . . . . . . . . . 156XV. Construction Management . . . . . . . . . . . . . . . . 161XVI. Start–Up Acceptance . . . . . . . . . . . . . . . . . . . . 162XVII. Project Turnover and Installation Qualification . . . . 163XVIII. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . 165

References . . . . . . . . . . . . . . . . . . . . . . . . . . 166

I. INTRODUCTION

The design and construction of active pharmaceutical ingredi-ent (API) facilities is an extremely complex and challengingundertaking. The time required to design construct and vali-date a facility to manufacture API products must be balancedagainst marketing and regulatory considerations. A firm maybe required early in the drug development process to startinvesting in new production facilities or enhancing existingcapacity so that a product can be produced for testing, andeventually for full-scale production to meet the marketdemand. An API manufacturer must develop a comprehensive

127

Page 143: Active Pharmaceutical Ingredients

process=facility design and construction execution strategy toensure achievement of all regulatory, cost, and market objec-tives for the compound.

The successful completion of a newAPI process facility is afunction of good engineering practices, sound constructiontechniques, and a well-planned and documented start-up andvalidation plan. Early detailed process definition enables theproject team to develop a comprehensive project executionstrategy. The execution strategy outlines the engineering andconstruction methods for the project. The start-up and valida-tion plan ensures regulatory compliance and a smooth transi-tion from construction to operation. Active pharmaceuticalingredient production facilities are complex, expensive todesign, construct, and validate. New facilities require sophis-ticated processing equipment, utilities, and support functions.Careful planning and good sound engineering is critical toassure that the investment in capital is managed wisely.

The reader must be cognizant of current good manufactur-ing practices (cGMPs) requirements for new products. Thedesign engineer will be responsible for design of facilities andsystems that will meet cGMPs for APImanufacturing. The con-structor will be required to install and validate the equipmentand facilities to meet the same criteria. Certain utilities, suchas process-deionized water, are required to meet specific regu-lated criteria. Details of validation and cGMPs are discussedin other sections of the manual. We will focus on the impact todesign and construction by validation and cGMPs.

Various strategies utilized in the engineering and con-structions of new production capabilities, whether they arenew facilities or renovations to existing capacities, arereviewed. A clear execution strategy and the need to under-stand the scope of the project are important components thatare reviewed in detail.Management of the design process is cri-tical to success. The firm must be able to properly manage thedevelopment of the design, ensuring that the process is com-plete and ‘‘frozen’’ prior to the commencement of construction.

Many of the terms and references in this section are typi-cal of fine chemical manufacturing. We will assume that thereader has an understanding of fine chemical manufacturing.

128 Mongiardo and Bobrow

Page 144: Active Pharmaceutical Ingredients

II. BUSINESS REQUIREMENTS

An API project is created when a need arises through one ormore of several areas. Some examples include: (1) new pro-duct introduction, (2) regulatory requirements, (3) existingproduct capacity shortfalls, and (4) process improvements.Any one or combination of these areas can generate the needfor a new capital investment.

The engineering and construction steps are similar for thefour stated cases. Excellent scope definition and a well thoughtout execution strategy are required for all of them. A firm willanalyze many different manufacturing options before estab-lishing the final project scope. The most uncertainty occurswith the new product introduction. The firm can be requiredto develop a preliminary scope of work during early stages ofprocess development. There is a higher probability of changeand process churn as the new process develops. The firm mustbe prepared to manage facility and equipment changes as thenew process is finalized. The key to success is both minimizingand coping with those changes. The uncertainty in the volumeof production requirements can also change during the initialscope development of a project.

The other business cases normally have a defined processwithin existing operating facilities and with known marketvolumes having already been on the market. The major pro-cess components have already been defined. The process is aregulatory agency-approved process, which has proven viabi-lity. Normally, the changes associated with these types ofprojects are limited in scope to process enhancements, i.e.,increasing throughput, eliminating bottlenecks, increasingyields, etc. We will not focus the discussion on these typesof projects.

We will focus on the requirements of engineering andconstruction of facilities for a new API introduction. This isthe most difficult and complex task because the technologyis untested on a commercial scale and there are technicalassumptions with the associated risks that must be taken.Assuming the product and the process are untried at commer-cial scale, there may be unforeseen issues with start-up and

Design and Construction of Facilities 129

Page 145: Active Pharmaceutical Ingredients

operation that arise at the proposed scale for the new APIentity. Certain components of the process, such as producthandling and transfer, and material consistency may becomean issue at the production scale, which were not detected in apilot or bench operation. The design team must take into con-sideration any components of the process that will not be ascaled-up duplication of the laboratory version of the process.The risk of a pump or product transfer system not workingproperly because of material viscosity or incompatibilitymay require changes to the process once the system is built.

Material handling aspects through equipment suchas centrifuges, blenders, or mills can be different from thesmaller-scale experience. Common problems that develop atcommercial scales include pumps not operating as designed,material bridging in centrifuges and blenders=dryers, anddifferent milling consistencies. The engineering and construc-tion team may be required to change components during theinitial production runs of a new compound. A good designerwill incorporate the necessary flexibility in the new processto allow for equipment change outs. The design of equipmentshould incorporate the ability to replace it or upgrade in amanageable fashion.

A benefit for the readers is the ability to utilize thisstrategy for other process improvement or regulatory-drivenprojects. The steps are similar if not the same (the major dif-ferences are associated with the business and engineeringanalyses for the new API).

A. An API Manufacturer Will Focus on theAppropriate Level of New Capacity

Market projections will indicate required volumes of the newAPI. Unfortunately, market projections can vary widely.Manufacturing capacity for a new API facility is expensiveto build, maintain, and operate. It is important to ‘‘properlysize’’ the production processing equipment and supportingfacilities. A proper engineering approach will incorporatethe ability to expand a production facility or equipmenttrain(s) to allow for future expansion for potential volume

130 Mongiardo and Bobrow

Page 146: Active Pharmaceutical Ingredients

increases. The future expansion planning can be as simple asincorporating the footings for a building expansion duringconstruction of a new facility. It can be as complex as addingan additional bay to a new or existing building along with allassociated utilities for that future expansion.

B. The New API Manufacturer Will Focuson Flexibility in Design

A key component of the analysis is whether to produce thenew API with dedicated process trains and facilities or tocampaign the new product with other products utilizing simi-lar equipment. Major new API compounds may warrant dedi-cated process trains and facilities due to the sheer volume ofproduct or due to unique processing techniques. However,most new higher-potency APIs can be produced with lessequipment over a shorter time span than in the past, thusallowing the manufacturer to produce multiple productswithin the same equipment. This approach can often resultin significant conservation of capital.

C. Location of the New Facility

The new facility can be located in any of the major markets inthe world. Many countries provide tax incentives for locatingan API production facility in their country. Labormarkets arean important component of the analysis. The technical skillsrequired to operate and maintain the facility and for construc-tion and start-up are sophisticated. Complex processes requireskilled technicians to run the process. Skilledmechanicswill berequired to maintain the facility.

Sophisticated equipment and facilities require skilledlabor and construction professionals. It is difficult to constructandmaintain one of these facilities in a remote part of theworldand certain parts of the United States. Labor markets are lim-ited. The new facility may compete with other facilities underconstruction for the available labor and construction supportresources. Major API and biotechnology projects have recentlyexperienced large cost impacts, both in the United States andforeign locations, because of a dearth of trained construction

Design and Construction of Facilities 131

Page 147: Active Pharmaceutical Ingredients

and engineering professionals to design and construct thesefacilities. Site selection should be one that offers an acceptablesupply of operational support and construction resources.

III. DEVELOPING THE PRELIMINARY SCOPE

A preliminary scope of the new process should be developed inparallel with process chemistry and engineering developmentand early piloting for the new API. The scope should include adefinition of the process, preliminary process flow diagrams(PFDs) and piping and instrumentation diagrams (P&IDs),equipment specifications and requirements (vessel types andsizes), preliminary facility fit, permitting requirements (localbuilding and environmental), and any regulatory (cGMP)requirements. The producer’s process-engineering group willhave to determine the best process fit to ensure speed to mar-ket, cost-effective manufacturing, compliance with safety andenvironmental requirements, and GMP compliance.

An analysis of alternatives is desirable once enough pro-cess definition is developed. Process siting and developmentdecisions should be based on scientific, business, and regula-tory analysis. Questions the producer should consider in thepreliminary scoping exercise include:

� Should the facility be multiuse (Fig. 1) (campaigning)or dedicated?

� Can the API be manufactured in an existing facility(retrofit) or will it require a new facility?

� Will the new=retrofitted facility be cGMP compliant?� What are the safety and environmental concerns of

the new compound?� Utilities:

– What is the status of process water and buildingutility systems?

– Do existing assets have the utility capacity(s) forexpansion?

� Schedule and cost: What is the best approach to sup-port a product launch?

132 Mongiardo and Bobrow

Page 148: Active Pharmaceutical Ingredients

A. Campaign vs. Dedicated

Many of the newer compounds developed for market are of ahigher potency, reducing the need for large (greater than 1million kilograms annually) volumes of the API. A dedicatedprocess may be the easiest approach to design and construct,but may not be the most cost effective or strategic. A dedi-cated process is ideal for a one-product organization or high-volume product. It may be easier to manage, with unchangingprocessing parameters. Varying market product demand canimpact usage of the facility and the cost of operations.

A campaign style facility will allow the manufac-turer to better utilize assets, integrating different product

Figure 1 Typical vertical processing facility designed for batchproduction with barrier separation between processing steps.

Design and Construction of Facilities 133

Page 149: Active Pharmaceutical Ingredients

manufacturing using similar equipment configurations. Theproducer has different options available for product volumesand production time. The campaigning facility will have differ-ent processing capabilities (Fig. 2) throughvariousmanifolds orhardpiped equipment configurations (equipment trains). Theseconfigurations can be manipulated for different processes. Thisprovides the manufacturer with the flexibility to vary produc-tion sequencing to produce several products vs. one.

Good manufacturing practice (GMP) considerations mustbe reviewed carefully with a multiuse facility. Good manufac-turing practices controls are applied with the use of APIstarting materials. The controls increase as process proceedsto final isolation and purification. The producer will

Figure 2 Typical specific ventilation system designed to protectoperators during the care of vessels with cytotopic-high potencycompounds.

134 Mongiardo and Bobrow

Page 150: Active Pharmaceutical Ingredients

be required to ensure GMP integrity for the new orrenovated facility. Some of the considerations include properproduct isolation (barrier separation), cleaning systemsfor multiproduct equipment (CIP—clean in place) andpharmaceutical grade water systems (for isolation andpurification).

B. New vs. Retrofit

The API manufacturer will be required to decide whether anew facility will be required or an existing facility can beretrofitted. Questions that the manufacturer should askinclude:

� What existing assets are available in the manufac-turer’s portfolio?

� Can these assets be modified to process the newproduct?

� What are the costs associated with the renovations?How do they compare to a new facility(s)?

� What renovations are required to qualify the processor facility?

A careful cGMP review will be necessary as part of ana-lyzing an existing facility for a new product fit. The currentguide for GMP guidance for API facilities is the InternationalConference on Harmonization (ICH) of Technical Require-ments for Registration of Pharmaceuticals for Human Useguide Q7A—referred to as ICH Q7A (1).

Facilities currently manufacturing fine chemicals maynot meet the standards outlined in ICH Q7A and not holdup under the scrutiny of a regulatory inspection. Majorrenovations may be required retrofitting an existing facility(s)to assure GMP compliance. The manufacturer can be requiredto install new systems such as CIP and pharmaceutical gradeprocess water. In product isolation=purification and finishingfacilities, the manufacturer will have to insure productseparation in multiproduct suites, through physical barrierssuch as walls, and also through differential room pressuriza-tion with minimum room air exchanges.

Design and Construction of Facilities 135

Page 151: Active Pharmaceutical Ingredients

Understanding the requirements of a GMP facility iscritical to developing an accurate cost and schedule modelfor the new product. A process fit that appears simple for a finechemical could require substantial renovations for an API.

C. Equipment and Facility GMP Compliance

Good manufacturing practice regulations affect the architec-tural and building engineering components of the buildingalong with equipment and systems. The building must be cap-able of providing items such as adequate lighting, properwaste water management, validated process water, productseparation areas (warehousing), and heating ventilation airconditioning (HVAC) and room separations for final step (iso-lation=purification) processing. The facility should have theappearance of a pharmaceutical facility. The processing areasshould be clean and free of debris. ‘‘Cleanability’’ is critical forall processing equipment involved in ‘‘critical step’’ and post-critical step manufacturing.

D. Safety and Environmental Concerns

Many of the new APIs are designed with a higher potency(cytotoxic) than previous generations. The stronger potenciesrequire the designer to integrate materials handling andHVAC systems that protect the operators from exposure tothe product. Specific ventilation systems are incorporated toprotect personnel while charging and operating vessels(Fig. 2). The facilities are designed to contain all materialswithin the confines of the facility. Similar to sterile processing,there will be air locks separating the different rooms (Fig. 3).

High potency facilities will normally have separate com-partments for gowning=dressing and entering, proces-sing=manufacturing, and decontamination=degowning. TheHVAC system will be dedicated for the facility. Wastewaterwill be discharged to a holding tank for testing prior to dispo-sal. The concept for the facility is total containment.

Process wastes will be managed similar to any organicfine chemical operation. The producer must separate andcontain all waste materials not suitable for wastewatertreatment.

136 Mongiardo and Bobrow

Page 152: Active Pharmaceutical Ingredients

IV. UTILITIES AND BUILDING SYSTEMS

A. Process Water Systems

Water systems are expected to be demonstrated to be suitablefor their intended uses. At a minimum, water is required tomeet the World Health Organization requirements fordrinking (potable) water. Processing steps such as isolationand purification will require purified water as outlined inUSP 23 (2), pharmaceutical grade water. Validation of watersystems is required for all product contact water systems. We

Figure 3 This high potencymaterials all lock provides separation ofmaterials from personnel and provides separate pressurized entranceand exit points. The high potency processing space will be negativelypressurized to the adjoining spaces effectively containing any airbornematerials exposed during processing. Operators enter through the airlock, change into protective gear, and enter the processing area. Thedoors are typically interlocked—not allowing someone to enter intothe processing area if the outer door is open. Once work assignmentsare completed, individuals exit through the degowning chamber,showering and removing contaminated outerwear before exiting.The shower water is contained in a holding tank for disposal. The unitis fully self-contained.

Design and Construction of Facilities 137

Page 153: Active Pharmaceutical Ingredients

will discuss design of purified water systems in more detaillater in this chapter.

B. Gases

Gases used in final processing steps will also require valida-tion and GMP compliance. These gases, such as nitrogen, willbe required to pass through filtration systems to remove anymicrobes that might be in the gas stream.

C. Heating Ventilation and Air Conditioning

During development of the preliminary scope, the engineershould take into consideration any HVAC control issues forthe new product. The design of the new or retrofitted facilitymust be cGMP compliant with respect to HVAC controls forall ‘‘final step=post final step’’ processing areas. Products thathave specific temperature or humidity requirements must bemanufactured in facilities that will assure regulators of thoseconditions for critical and postcritical processes. Dry proces-sing steps such as milling, drying, and blending are to be per-formed in areas that assure the manufacturer and regulatorsof no product cross-contamination.

V. PRELIMINARY SCOPE DELIVERABLES

The preliminary scope should include enough information forthe engineer and constructors to start developing cost andschedule data. The information should include process flowdiagrams, preliminary P&IDs (Fig. 4), initial facility require-ments, and the first cut at a validation strategy.

A. Contracting Strategy

Once the API manufacturer has generated enough initialinformation for the new process, they will have severaloptions for implementation of the design and construction ofthe new facility. There are various execution strategies,which include:

� design and construction utilizing an engineering firmand a construction contractor(s);

138 Mongiardo and Bobrow

Page 154: Active Pharmaceutical Ingredients

� design and construction utilizing an engineeringfirm(s) and a construction management firm(s);

� utilizing one firm to provide engineering, construc-tion, and procurement services.

The use of the one firm concept of engineering, procure-ment, and construction is well suited for a manufacturingfirm that does not have depth within its own engineeringand procurement organizations. This method places all theresponsibility on the engineering contractor to deliver a fin-ished GMP compliant facility. The firm selected must havethe personnel depth to be able to supply all facets of the pro-ject. This method tends to be the most effective for schedule,but can carry cost premiums. The engineering, procurement,and construction (EPC) contractor assumes all the risks onthe project and will charge a premium for assuming the risk.

Figure 4 Example of a P& ID for process utilities system (cleansteam generation).

Design and Construction of Facilities 139

Page 155: Active Pharmaceutical Ingredients

The other methods work better with manufacturingfirms that have established plant and construction engineer-ing groups. The first method, design and contract, is the‘‘traditional’’ method of construction. The design is completedand the project bid. The successful bidder has a lump sumcontract to complete the work. This method tends to takelonger. However, in a competitive market it can be the mostcost effective. Utilizing a construction manager and separatedesign, firm will enhance the schedule of the project by bid-ding work as it is designed and will control costs if properlyexecuted. The chapter discusses these methods in detail later.

B. Development of the Design Strategy andDetailed Design

The API manufacturer has essentially two choices in setting adesign execution strategy. They may elect to develop thedetailed design in house with their own expertise or they willobtain the services of an engineering firm=contractor who willprovide the services for them. We will discuss the option ofutilizing outside services for this function. The vast majorityof manufacturing firms do not possess the ‘‘in-house’’ capabil-ities to develop the full breath of design for a new processfacility or a major renovation.

There are various methods of employing the outside firmutilizing various contracting strategies. The firms can be hiredon a reimbursable basis. This method is the most common inthe industry. The firm is remunerated for all design servicescost plus a mark-up for overhead and profit. Typically, themanufacturer will negotiate a contract with the designservices supplier for a ‘‘not to exceed’’ value for the work. Thedesign firm normally develops this estimate. It is impactedby the stage of process development. Themore defined the pro-cess and the scope of work, the better the estimate.

The other less common method is buying the design on alump sum basis. The design firm provides a firm price for thework. A careful definition of expectations is required for thisapproach. This contracting strategy is akin to using a build-ing contractor for a home or commercial building. The pricing

140 Mongiardo and Bobrow

Page 156: Active Pharmaceutical Ingredients

is based on a fixed set of parameters, which are normally theplans and specifications developed for project. Any items notincluded in the plans and specifications are considered outof scope. All items out of the scope of the contract are subjectto extra charges. The lump sum design contracting strategy isa difficult strategy for designing a new process with uncer-tainty. As design progresses, any changes to the process willresult in negotiating change orders to the contract with thedesign firm. This method can create distractions to the designeffort as the manufacturer and the engineer become involvedin pricing negotiations. This method is more common withsmall process configuration changes in existing facilities.

Selecting the right firmand establishing clear expectationsis critical to the success of the project. How the firm is utilized isa decision to be made in the planning stages of the project. TheAPI manufacturer can elect to use the various methods of con-tracting for services that have been outlined in the chapter.

C. Setting Expectations

The execution of the design and construction process for anAPI facility can be defined in four critical steps:

1. design2. procurement and construction3. equipment validation4. start-up, commissioning, and turnover

We have briefly discussed different contracting options.The API manufacturer has to decide how to procure theoutside services necessary to accomplish these steps. We can-not recommend any one method as better than another. Thecontracting decisions must be made based on all party’s rela-tive strengths in the execution of this type of project. If thedecision is to contract out the entire process to one firm inan EPC contract, the outside firm is expected to deliver a com-pleted facility, validated and ready to produce product.

The API manufacturer, with some level of in-houseexpertise, can elect to manage the design and constructionseparately. The outside design firm will be responsible for

Design and Construction of Facilities 141

Page 157: Active Pharmaceutical Ingredients

providing the proper level of documentation for a constructionfirm to execute the work. The API manufacturer exercises agreater level of control in this process. The API manufacturerwill be involved in many of the decisions made in procurementof equipment and other components and be better able toinfluence the operability of the facility.

VI. DESIGN DEVELOPMENT

The design of a new API facility will develop from an initial‘‘napkin’’ exercise to a set of documents that a constructor willuse to install the new assets. Progression of the design can beinferred from the following sequences: preliminary scope, basisof design, and detailed design. Many companies in the petro-chemical and chemical industries utilize this practice. Recently,this progression has been utilized in the API industry.

Development of the design is a function of process defini-tion. Once the process is clearly identified, the API manufac-turer and=or the design firm can complete the in-depthanalysis of existing assets and new assets to progress the designfor the project.

As previously discussed, the preliminary scope definesthe major components of the API facility. The scope will haveidentified key processing steps, all associated equipment, andany facility requirements. The preliminary scope will be a keydocument in communicating to outside design firms the intentof the facility(s) and the overall process intent of the project.The API manufacturer must then decide how they want tomanage critical steps of the design, construction, validation,and start-up process.

Another phase in design development currently utilizedin the chemical=petrochemical industry is a ‘‘basis of design’’phase. The basis of design has certain components of the designdefined prior to a final cost estimate and schedule is completed.The basis of design has components such as ‘‘approved fordesign’’ P&IDs, PFDs, and facilities definitions already deter-mined. The basis of design will outline: permitting require-ments—local government and environmental, engineeringcriteria for the new site (civil and infrastructure), and a preli-

142 Mongiardo and Bobrow

Page 158: Active Pharmaceutical Ingredients

minary project execution strategy. Typically, the basis ofdesign represents approximately 20% of the total design.

Once the basis of design is completedand thefinal estimategenerated, the design team will develop detailed design docu-ments, which will incorporate all the necessary informationfor the builder(s) to construct, and start-up the facility(s). Thefollowing components should be defined during this process:

� equipment requirements� facilities requirements� utilities requirements� safety requirements� cGMP requirements� qualification or validation plan� expansion capabilities� hazard and operability (HAZOP) analysis� process and instrumentation diagrams (P&IDs)� enviornmental requirements (permitting)

A. Equipment

The P&IDs will identify the major equipment components tobe modified or procured for the new process. It will outlinethe new equipment and associated controls required to runthe process. The API manufacturer may have preferredsuppliers of this equipment because of operability or main-tenance issues. Most of the vessel and component suppliersin the industry are capable of supplying cGMP compliantequipment. The design firm or the API manufacturer willspecify the equipment to include the necessary appurtenancesto make the equipment cleanable.

The design firm normally generates procurement specifi-cations. These specifications will include definition of allmajor components such as materials of construction, agitatorrequirements, nozzles, etc. Major equipment for API proces-sing is similar to fine chemical production and can includereactors, centrifuges, condensers, heat exchangers, distillationcolumns, extractors, absorption equipment, chromatographyequipment, dryers, blenders, crystallizers, mills, etc. Thesecomponents will normally require validation [installation

Design and Construction of Facilities 143

Page 159: Active Pharmaceutical Ingredients

qualification (IQ)=operational qualification (OQ)=performancequalifications (PQ)] prior to manufacturing. It is important tohave proper coordination between the design engineer and thevalidation team. The validation team will be responsible forgenerating validation protocols developed from the equipmentspecifications and processing parameters.

B. Facilities

Many of the recent cGMP initiatives have been focused onfacility requirements. The manufacturer will be responsiblefor adhering to these requirements for processing, productseparation, materials handling, and utilities. Because of theserequirements, additional space for functions such as ware-housing may be required. ICH Q7A is the document the pro-ducer will refer to for information on facilities and processingrequirements. The new facility will require enough space toprovide separation of raw materials (i.e., quarantined vs.approved) and for finished products and intermediates (quar-antined vs. approved). The manufacturer must be able to iso-late and segregate these components.

Manufacturing space, containing equipment involved incritical and postcritical step processing, is required to main-tain a level of cGMP compliance consistent with the stage ofthe product. There are noticeable differences between inter-mediate facilities and facilities that manage final (criticaland postcritical) steps of an API process.

An intermediate facility will not require the level of sop-histication normally associated with a final process facility.There will be differences with respect to equipment separation,architectural finishes, utilities, and general building configura-tion. Some of the differences include less costly equipmentseparation (open bays vs. rooms); architectural finishes consis-tent with fine chemical manufacturing, and potable (drinking)water vs. purified water.

The isolation=purification facilities will be designed withGMP considerations consistent for the final stage of the pro-duct. Some of those considerations include product isolationand separation (Fig. 5), cleanable surfaces, purified water sys-tems, and temperature, airflow, and humidity controls.

144 Mongiardo and Bobrow

Page 160: Active Pharmaceutical Ingredients

C. Product Separation

Product isolation is required for processing facilities involvedin critical and postcritical processing. Production facilitiesinvolved in campaigning different products must be able toprovide adequate separation for product isolation duringtransfer of product. The producer must be able to assure theregulatory agency that all products are produced in an envir-onment free of the potential of cross-contamination. This iso-lation can be performed through the use of temporary barriers

Figure 5 This figure represents a self contained product transfer‘‘box’’ that allows product to be transferred from a piece of equip-ment such as a centrifuge to a drum protecting the product fromcontamination. The unit also provides operator protection from highpotency compounds.

Design and Construction of Facilities 145

Page 161: Active Pharmaceutical Ingredients

such as curtains or through the construction of separate bayswith permanent physical barriers (walls).

Isolation for final processing steps is best accomplishedthrough the design of individual rooms (suites). The roomsare finished with smooth, cleanable, durable finishes suchas epoxy. All utilities are piped in from adjacent mechanicalrooms. All penetrations are sealed to maintain the properenvironment in the process suite. The vessel heads and manways should be kept free from overhead components thatcould collect dust. Heating ventilating air conditioning sys-tems are designed to maintain positive room pressurizationwith respect to connecting corridors. This will protect theroom from contamination by dust or other particulates. TheHVAC system will have particle filtration that will filter outairborne contaminates.

Sterile processing facilities require additional levels ofsophistication. Active pharmaceutical ingredients manufac-tured for sterile use are required to be completed (usuallythe isolation=purification steps) in a sterile facility. The ster-ile facility is designed to minimize the exposure of the productfrom microbial contamination.

CFR 211.42 (3) states: (design and construction features)requires in part, that aseptic processing operations be ‘‘per-formed within specifically defined areas of adequate size.There shall be separate or defined areas for the firm’soperations to prevent contamination or mix-ups.’’ Asepticprocessing operations must also ‘‘include, as appropriate, anair supply filtered through high efficiency particulate air(HEPA filters) under positive pressure,’’ as well as systemsfor ‘‘monitoring environmental conditions . . . ’’ and ‘‘maintain-ing any equipment used to control aseptic conditions.’’

Section 211.46 (ventilation, air filtration, air heating,and cooling) states, in part, that ‘‘equipment for adequate con-trol over air pressure, microorganisms, dust, humidity, andtemperature shall be provided when appropriate for the man-ufacture, processing, packing or holding of a drug product.’’This regulation also states ‘‘air filtration systems, includingpre-filters and particulate matter air filters, shall be usedwhen appropriate on air supplies to production areas.’’

146 Mongiardo and Bobrow

Page 162: Active Pharmaceutical Ingredients

The building or suite is designed with separation andcontrol. Air quality will vary depending on the nature of theoperation. The area design is based upon satisfying microbio-logical and particulate standards as defined by the equipment,components, and products exposed, as well as the particularoperation conducted, in the given area.

There are two clean areas that are important to sterileAPI product quality. The critical area (class 100) and thesupporting clean areas associated with it.

Class 100 conditions require that air in the immediateproximity of exposed product be of an acceptable quality witha particle count of no more than 100 0.5-mm particles per cubicfoot of air. This is obtained by utilizing HEPA filters and lami-nar flow conditions with the room HVAC. Room pressuri-zation is also critical. Class 100 rooms are required tomaintain a positive pressure to surrounding rooms of at least0.05 in of water will the doors closed. The supporting cleanrooms can vary from class 1000 to class 100,000 dependingon the function with class 100,000 as the least critical to class10,000 for adjoining rooms.

The sterile processing buildings are designed with a hier-archy of separations. Manufacturing is separate from ware-housing, warehousing and manufacturing from offices andlocker facilities, and also separations from utilities. Materialflow is critical for a successful design of aseptic facilities.The designer should make every attempt to design the facilityfor unidirectional flow of components, ingredients, and pro-duct. Unidirectional flow means that materials and productall flow in one direction as the product steps through variousphases of completion. This provides the greatest assurancethat sterility will not be compromised as it might in a facilitythat did not possess unidirectional flow.

VII. UTILITIES

Utility requirements for API manufacturing are not atypicalfrom fine chemical manufacturing. Process equipment willrequire steam, water (potable, chilled, tower=cooling), nitro-

Design and Construction of Facilities 147

Page 163: Active Pharmaceutical Ingredients

gen, and vacuum. Jacket services can be designed for multiplefluid use or for single fluid applications, utilizing a multipur-pose fluid for both heating and cooling. Buildings should bedesigned with utilities separated from the processing areas.Chillers, heat exchangers, pumps, etc. should be located inseparate rooms, floors, or areas.

A. Water Systems

Water systems used in the manufacturing of APIs are subjectto validation guidelines as outlined elsewhere in this book.The manufacturer will be required to utilize a water systemas appropriate to the process. The United States Pharmaco-poeia outlines minimum specifications for various levels ofwater purity. The manufacturer will determine the qualitylevel required at all the various stages of molecule develop-ment. The manufacturer will utilize drinking water quality,potable water, for all precritical processes (intermediate steps)and purified water for critical and postcritical applications.

Purified water systems (Fig. 6) for pharmaceutical pro-cessing require a level of sophistication not required in theproduction of fine chemicals. There are many different waysto generate purified water. Following is a brief descriptionof a typical pharmaceutical grade water system. The systemwill include the following components:

1. treatment2. sanitization3. storage and circulation

1. Treatment

The equipment required for water treatment will be deter-mined by the quality of the incoming water. Typically, a USPpharmaceutical grade system will require pretreatment(filters), deionization, reverse osmosis, and potentially a pol-ishing step such as continuous deionization. Many systemsnow incorporate UV filters for sanitization, which kill micro-bials and also eliminate ozone.

148 Mongiardo and Bobrow

Page 164: Active Pharmaceutical Ingredients

2. Sanitization

The systems are designed to be cleaned. Recent industrypractices have included the use of ozone injected into thesystem as a sanitization step. Other methods include steamsanitization. The ozone is then eliminated through the UVfilters. The system should be designed for a complete sanitiza-tion, which includes all storage tanks and distribution piping.

3. Storage and Circulation

Typical systems include a storage tank with sanitization cap-abilities. Treated water is sent to the tank and then circulatedto the points of use in the manufacturing areas and returnedin a continuous loop. The circulation loops are normallydesigned to maintain flows that will inhibit bacterial growthwithin the distribution systems. The system designer mustbe aware of the minimum velocity requirements for these sys-tems. Typically, the designer will use a rule of 4–6 fps (feetper second) as a minimum velocity.

Figure 6 Example of a circulating purified water loop utilizingreverse osmosis, deionization, ultra violet lighting, and microfiltration.

Design and Construction of Facilities 149

Page 165: Active Pharmaceutical Ingredients

4. Materials of Construction

The system will be constructed of sanitary materials. Allfittings will be sanitary grade. Typically, these systems arebuilt utilizing PVC or stainless steel (316L), or a combinationof the two.

Other systems impacted by cGMP requirements (productcontact) include nitrogen and plant air. These systems willrequire filtration systems to insure no impurities are passedthrough and make contact with product.

VIII. SAFETY

As previously stated, process and facilities designs areimpacted by the potential need to include handling ofhazardous (cytotoxic) compounds. The newer higher potencycompounds are potentially toxic in the large volumes theyare produced.

Heating ventilation air conditioning systems for finish-ing facilities where potent=toxic compounds are handled arerequired to work under negative pressure. High efficiencyparticulate air or 95% air filtration systems are utilized toremove particulate from the air stream. In intermediate pro-cessing, isolation chambers may be required to protect thefacility environment. These chambers will contain the equi-pment that holds the material (i.e., centrifuge or dryer)and provide a physical barrier (plaster or block walls) andan air bath (under negative pressure). Operators working inthis environment will be required to wear personal protectiveequipment (PPE). The facilities will be designed to minimizethe hazard by limiting exposure to the individuals and envir-onment.

IX. CURRENT GOOD MANUFACTURINGPRACTICES REQUIREMENTS

Current good manufacturing practices requirements havebeen discussed throughout this section. The reader must

150 Mongiardo and Bobrow

Page 166: Active Pharmaceutical Ingredients

be aware of the requirements of ICH Q7A—good manufactur-ing practices for API facilities. This document provides guide-lines for manufacturing facilities for API products fromintroduction of the API starting materials through physicalprocessing and packaging.

X. QUALIFICATION PLAN

The qualification plan for an API facility consists of thefollowing:

� commissioning� validation� installation qualifications� operational qualifications� performance qualifications

The supplier of engineering services can also providecommissioning and validation services for the API manufac-turer. There are also third parties specializing in commission-ing, validation, standard operating procedure (SOP) writing,and operational training. Early in the project developmentprocess (initial scope development) the contracting strategyfor validation=start-up services should be determined. Engi-neering, procurement, and construction contracts can includethis as part of the suppliers’ scope. The API manufacturer willhave to decide how this work will be executed. The API firmcan perform the work internally with engineering resources.They can use the engineering firm of record or the contractorto perform the service—or they can hire a third party specia-lizing in validation=commissioning.

The engineering firm will be responsible for identifyingkey supplier documents required for validation (IQ and OQ)and also specifications and ranges for equipment. The valida-tion services will develop protocols for executing each compo-nent (IQ and OQ). Performance qualifications are performedafter completion of OQ. The engineer or contractor will typi-cally not be involved in PQ. Our experience has been thatthe producer will perform PQ on the new process.

Design and Construction of Facilities 151

Page 167: Active Pharmaceutical Ingredients

The IQ protocols are designed to verify that the installa-tion has been completed as specified. As an example, an IQprotocol for a vacuum pump will ensure that the right pumpwas installed, as specified. The entire nameplate data willbe recorded, documenting all necessary engineering informa-tion such as size, type, and purpose. All electrical and instru-mentation contacts will be tested and verified.

The OQ protocols will test all critical parameters for theequipment. It will test all control devices, calibrate criticalinstruments, and test major vessels under operating condi-tions (pressure and vacuum).

XI. EXPANSION CAPABILITIES

Earlier in this chapter, we identified the importance of ‘‘cor-rectly sizing’’ the process. Market projections for API pro-ducts can swing wildly. A recommendation for the reader isto be cognizant of the potential for expansion. When design-ing the new process facility or upgrading an existing plant,the API producer should position the plant to be expandedif necessary. Consideration should be made for preinvest-ment of some facilities or utilities during the design of thefirst phase. It is clearly cost effective as compared to havingto reinvest later. However, it is ultimately a managementdecision on managing risk. As previously mentioned, certainfacility components are easily installed at one time vs. stag-gered. A good example is piles and foundations. If the produ-cer anticipates product growth, the scope may include thenecessary subsurface components for future expansion.When the producer decides to expand some time in thefuture they will be able to avoid costly foundation excava-tions and associated disruptions of heavy subsurface civilconstruction.

XII. HAZARD AND OPERABILITY ANALYSIS

At various stages of the design process, a HAZOP (Fig. 7)must be conducted on the project. The purpose of this analysis

152 Mongiardo and Bobrow

Page 168: Active Pharmaceutical Ingredients

is to identify any potential weakness in the design of a processfacility. Weaknesses are identified as:

� safety concerns (i.e., dangers to personnel)� enviornmental impact (i.e., chemical release)� economic impact (i.e., damage or loss of equipment

or facility)

The HAZOP reviews will look at each detail of the process,examine what is happening in that stage of the process, andthen question a series of ‘‘what if’’ potential failures. Questionssuch as a failure of a control, loss of power, will generate a list ofpossible reactions to that failure mode. The failure list is thengenerated from experience with similar process arrangements

Figure 7 This sample HAZOP report identifies several hypothe-tical operating hazards that can occur during processing/operations.The report includes recommendations and safeguards assigned tospecific individuals for execution.

Design and Construction of Facilities 153

Page 169: Active Pharmaceutical Ingredients

or from experience with this specific equipment. This potentialfailure is then analyzed and a determination of the risks. A fail-ure of low risk from safety exposure or cost of damage to thefacility would not generate further action by the design team.An item with a potential of extremely unsafe condition or highcost damage would then be listed with a recommendation foradditional controls or a revision to the design.

XIII. EXECUTION STRATEGY AND PLANNING

In any project, the cost and schedule predictability is impor-tant. All projects, from an office building to a major manufac-turing facility, have external factors, which are affected bythe cost and completion of a project. When a project is firstconceived, the drivers for the project establish the cost andschedule goals. If a project is purely financially driven thencost is the major controlling factor. If speed to the marketdrives the project, then schedule becomes of paramountimportance. In general, the project execution strategy musthave a primary goal. If the project is falling behind schedulea decision must be made to determine if the cost of overtimeis justified. If cost is the driver then a slip in schedule maybe acceptable or, conversely, the cost of working overtimemay be justified to maintain a critical schedule.

The ‘‘project execution strategy’’ must be aligned with theoverall project goals. To align these goals we must firstunderstand how executing a project can support these goals.If cost is the main goal of a project, such as in commercial orgovernment projects, the contracting strategy is to have com-petitive lump sum bids submitted by as many general contrac-tors as can demonstrate the financial strength to complete theproject. This financial strength is demonstrated through thesubmission of a bid bond with the proposal. The quality of theend product is ‘‘industry standard.’’

This method of lump sum bidding requires all the designdocuments to be completed and the entire project constructionis awarded to one general contractor. This contractor will beresponsible for the procurement of the individual trade sub-

154 Mongiardo and Bobrow

Page 170: Active Pharmaceutical Ingredients

contracts and the procurement of all the equipment. Becausethe design is completed, and the basic coordination of thetrades has been engineered in advance of the start of con-struction, the extent of extras on the project is usually limitedto unknowns. Items such as subsurface conditions or, inthe case of renovations, existing conditions, which are notanticipated in the design, may be uncovered during theconstruction activities.

The administration=management of a lump sum generalcontract is also limited due to the completion of the designprior to the start of the construction activities. This projectapproach will take the most overall schedule time, but thefinal cost is easily predictable and known with great accuracyprior to the start of construction. The delivery of long leadequipment and fabricated materials such as structural steelwill often not support the overall project schedule. There willbe times in the construction schedule when the project is wait-ing for materials and equipment to be delivered. In the con-struction of most API facilities, schedule is an importantdriver of a project. The lump sum general contract approachwill not normally satisfy the schedule requirements of theproject.

Schedule time is normally reduced through a techniqueknown as ‘‘fast tracking.’’ Major equipment bid packages andearly construction packages (foundations, structural steel,etc.) are awarded prior to completion of the entire design. Asthe design progresses, other packages are released for bidding.A major component of the ‘‘project execution strategy’’ is thebidding strategy for the project. The system of fast track isnormally applied to the entire project execution strategy.Almost all API manufacturing projects are schedule driven.

Cost predictability for fast track projects is less definitive.The potential exists for early construction packages to haveerrors or omissions that would have been uncovered if theremaining design had been complete. Whenever the designis not complete there is cost uncertainty. Any late changescan impact design components that have already been orderedor constructed. Accordingly, the administration of a fast trackproject requires extensive field effort and manpower. The

Design and Construction of Facilities 155

Page 171: Active Pharmaceutical Ingredients

number of construction contracts increase and the complexityof the overall project increases. Most API manufacturing com-panies (the project owners) do not have the in-house resourcesto effectively administer a fast track project. The ownerstypically use resources from a construction management orengineering company to monitor field activities, procurement,cost, and schedule.

XIV. PROCUREMENT STRATEGY

There are various ways of contracting for construction anddesign management. An EPC or design build contract is thejoining of the engineering design, procurement, and construc-tion management functions with one supplier. Owners canelect to hire firms with the in-house expertise to manage allthree functions or a design firm and a construction manage-ment firm who have joined forces to provide these servicesunder one contract umbrella. The advantage of the single con-tract is the single point of responsibility between the ownerand the supplier.

Some owners will elect to have separate contracts withthe design firm and with the construction management firm,with the construction firm responsible for the majority of theprocurement. The advantage of separate contracts is thatthe owner retains control over the design, and the design workcan start before the construction management firm is selected.

When selecting a firm to manage the design and=or con-struction of the project, there are a number of strategy andplanning tools that these firms must use to effectively managethe overall process. During the various phases of the project,cost estimates and project schedules are prepared. Theseestimates and schedules are constantly refined as moredetails of the project are developed.

At the conceptual stage of a project, the cost estimate(s)developed are utilized for the project’s overall business strat-egy. The initial return on investment (ROI) calculations arebased on these preliminary estimates. These estimates andschedules although based on limited data, usually not more

156 Mongiardo and Bobrow

Page 172: Active Pharmaceutical Ingredients

than 10% of the total engineering, are critical to achieving theproject’s overall goals. The selection of the key firms to supplythe design, construction management, and estimating ser-vices is a critical early project activity. The preparation ofthe project estimates is usually a collaboration between theoutsourced vendors and contractors who are familiar withthe construction and design market, and the API manufactur-ing (owner) who is more familiar with the chemistry or theprocess and the cost structure of the company.

As the design progresses the estimate is refined. Theassumptions made during the conceptual estimates must beevaluated for changes. In the initial conceptual estimate,there were constructability assumptions used to prepare theestimate, such as labor availability, equipment and materialdeliveries, and the sequence and methodology of the construc-tion work. At each refinement, the level of uncertainty isreduced and therefore the level of contingency for unknownsis also reduced. The overall contingency required for a projectis in direct relationship to the level of uncertainty or predict-ability of the final cost of the project. In the initial stages ofthe project (10%), a contingency of 25þ% is common. At thecompletion of the detailed design and with the process com-pleted, the contingency should not be required at greater than10%. In a complex process, an additional contingency is estab-lished for the final start-up and validation activities. The levelof risk, which drives this contingency, is based on the com-plexity of the process.

When a project is ‘‘schedule driven,’’ it is imperativethat a schedule be established early in the project and beutilized during the project. During the project planning,the schedule will grow in complexity and task breakdownas the overall project is developed. The schedule must be aworking plan throughout the project. This plan will beupdated as new data are known and to reflect the currentapproach to the overall project execution. An effective sche-dule must have relationships between the work items. Thesize and number of schedule activities on a project vary fromproject to project. Many computer programs exist that canarrange the schedule data in easy to analysis formats. A pro-

Design and Construction of Facilities 157

Page 173: Active Pharmaceutical Ingredients

ject critical path method (CPM) schedule, to be effective,must have the necessary detail to show a clear criticalpath. The bar chart is the most common display of a projectschedule (Fig. 8).

Figure 8 A typical bar chart.

A schedule can also be produced in an arrow diagram,which will graphically show all the activity prior to or depen-dent on an activity and subsequent activities, those, whichfollow an activity. This presentation can be very useful inthe analysis of how a project can be executed. To monitor aproject schedule effectively, the level of activity should bedetailed to show the items that should be accomplished dur-ing a specific period of time to maintain the overall projectcompletion schedule. The capabilities of the design andconstruction management firms or EPC firm to produce andmonitor an effective project schedule in the complexityrequired to manage these projects is an important elementin the selection process of those vendors.

The selection of the architectural=engineering (A=E) andconstruction=management (C=M) firms and the contractingstrategy with those firms is a function of the schedule drivers.Activities must be worked on concurrently to support the

158 Mongiardo and Bobrow

Page 174: Active Pharmaceutical Ingredients

schedule. The suppliers need to provide resources for projectplanning, early long lead procurement, and conceptualestimating. In many cases, the early involvement of thesesuppliers is contracted on a reimbursable or cost plus basis.As the project becomes better designed and scoped, the con-tract between the owner and the A=E and C=M supplierscan become a guaranteed maximum price, lump sum or areimbursable contract with schedule, and cost incentives. Inplanning the execution strategy, the resources for the start-up and validation must also be identified early in the process.Many engineering firms have the in-house resources to planand manage the start-up and validation activities. This is alsoimportant to decide when selecting the overall procurementstrategy for the project.

With an effective, realistic cost estimate and CPM sche-dule in hand, the manager of a project can make effectivedecisions regarding the planning and execution strategy.Many times marketing decisions will dictate the project com-pletion date, which could require additional funds to allow anacceleration of the project by either working overtime or add-ing additional shifts. When evaluating the final schedule foran API project, the time required for the start-up and valida-tion of the facility is critical to the success of the project.These activities usually start at the completion of construc-tion; however, their duration and requirements make itnecessary to start these activities when phases of theconstruction are complete. Overlapping of these activities willalso reduce the overall project schedule. The early planningand strategic development of an overall project strategy willidentify schedule opportunities.

The sequence of the construction can then be planned tosupport the validation. The development of the validationstrategy should be developed as part of the overall projectexecution planning. The strategy for contracting for the vali-dation services is a critical early activity. It is important toidentify the process systems that affect or come in contactwith the product. These systems must be validated. In a pro-cess if chilled water or steam is used to heat the jacket of avessel but that steam or chilled water never comes in contact

Design and Construction of Facilities 159

Page 175: Active Pharmaceutical Ingredients

with the final product, the utilities will not usually requirevalidation. However, the instruments that control the steamto the vessel jacket will usually require validation. If thecontrols do not function properly then the product can beoverheated or cooled. All pipe systems that transport productmust be totally validated.

Another element critical to the completion of a project isthe time required for testing of equipment and control systems.In the project validation and procurement planning, all equip-ment, and systems, which require factory acceptance testingprior to shipment, should be identified. The specifications,and procurement documents should provide for the requiredtesting and identify documentation of the testing procedurefor the equipment and control systems prior to shipment tothe site. In many cases, with skid-mounted equipment thatoften has microprocessor controllers, a significant amount ofthe IQ documentation and verification can be accomplished atthe factory. This preplanningwill save the overall project sche-dule. The testing, documentation, and validation activities arethen accomplished in parallel with construction activities.Equipment problems are flushed out at the factory prior tothe installation in the field. This pretesting at the factory canincrease the productivity of the construction installation andreduce the overall project schedule.

In the development of the process automation and con-trol system, the required testing of that control system andthe factory-assembled components, and the process simula-tion program must be established with the general func-tional specifications. In an API facility, many of thecontrol systems perform process functions that require strictvalidation. The functional description for the automationsystem should require a complete factory acceptance test(FAT). This test should simulate the entire process and pro-cess failures and alarms. The FAT should also check andverify that the control system cabinets and controllers oper-ate as designed. The factory acceptance testing of the pro-cess automation system prior to shipment and installationin the field is a critical step in the validation and start-upof the facility.

160 Mongiardo and Bobrow

Page 176: Active Pharmaceutical Ingredients

XV. CONSTRUCTION MANAGEMENT

As the project transits from the design stages to the construc-tion phase of the project, the construction management planand subcontracting strategy are developed and finalized.The subcontracting strategy has been completed early in theoverall construction plan. Design documents must be pre-pared to support the schedule of the construction activitiesand the submittal of vendor drawings and documents isnecessary to complete the design effort. Consistent withequipment, early contracting of the process automation sys-tem will start the submission of vendor drawings to the engi-neer for both approval and for inclusion into the electrical andinstrumentation drawings.

There are many components to the facility, which maynot have a long delivery but may be critical to the completionof the various design and construction packages. Controlpanels, IO cabinets (the termination cabinets for the inputand output wiring of instrumentation) and uninterruptedpower supply (UPS) systems need accessibility, which canaffect the architectural layout of support space and controlrooms. Instrument details are needed from the vendors tofinalize the electrical drawings and pipe fabrication drawings.Many times these smaller components will be included in thecontract to be purchased by the subcontractors, rather thanthe API manufacturing companies (the project owners). Theconstruction management firm and the design firmmust coor-dinate these details to insure that the information necessaryto complete the design is passed from the vendors to thedesigners to support the overall project schedule.

The development of a detailed construction schedule isnecessary for the coordination of the various contractorsand monitoring the progress of the construction. A resource-loaded schedule for construction activities is necessary. Laborrequirements must be evaluated from both availability andthe density within the construction project. A general ruleof thumb is to plan on one construction worker for every200 gross square feet of building. In the case of compact pro-cess facilities, this can require spot density of one worker

Design and Construction of Facilities 161

Page 177: Active Pharmaceutical Ingredients

every 150 ft2. In the electrical and instrumentation trades,this may be as tight as one worker every 100 ft2 of gross floorspace. The ability to fabricate pipe skids and systems, offsite must be considered both to control labor density and tosupport the overall schedule goals. The construction schedulemust be detailed enough to be able to show the different criti-cal paths but simple enough to be understandable by thevarious trade workers.

The construction manager’s involvement during designdevelopment is critical for constructability reviews to becompleted at all phases of the design. There is a constantreview of how the facility will be built and whether or notthe design is practicable. Typical questions include: in whatsequence will the facility be constructed? Does the designallow the different trades to complete their work withoutblocking other trades? Any contractor, who must remobilizeto complete their work, will add schedule to the project andcost.

Final documentation for a validated manufacturingfacility is critical to the success of the project. Documenta-tion and certification of the work is the responsibility ofthe construction management team. The format and itemsrequired for certification should be included in both thedesign documents and the validation plan. Almost all docu-mentation provided by vendors is available in electronic for-mat. The computer design programs used to provide thisdocumentation should be established in the early stages ofthe project and validation plan. The construction managerinsures that the subcontractors and suppliers comply withthese requirements. Weld documentation and instrumentcalibration are two of the most common certificationsrequired of the contractors.

XVI. START–UP ACCEPTANCE

The start-up and operational acceptance of any process facil-ity is a complex undertaking, which requires early planning.As systems are started and functioning, they can be reviewed

162 Mongiardo and Bobrow

Page 178: Active Pharmaceutical Ingredients

with the operation personnel. Training of the operationspersonnel for a process facility should start during the finalstages of the construction. Normally, training and fieldstart-up support is included in the purchase contracts for allmajor equipment and system suppliers. The training consistsof classroom theory of the equipment or system, the requiredmaintenance, and their intended functions. Classroom train-ing should occur prior to completion of installation of equip-ment. The operations personnel will then be familiar withthe equipment and have participated in validation of thesystems.

In an API facility, the construction manager is taskedwith coordinating the overall construction and installationof the equipment within the process facility. The installationof the interconnecting components must be installed in aquality manner. Rework takes time and delays the entireprocess.

XVII. PROJECT TURNOVER ANDINSTALLATION QUALIFICATION

The installation qualification (IQ) of a facility is the verifica-tion that all the components within a facility that have‘‘direct product impact’’ are installed correctly and in accor-dance with the design specifications. There must be support-ing documentation that all components have been installedand that all instruments have been properly calibrated.The validation plan defines the methodology for preparingand executing the IQ. It also provides guidance with respectto IQ acceptance criteria, the use of support documentation,and responsibilities. The construction and validation teamshould perform a walk down of the completed system. Thewalk down is a review of the completed installation of the con-struction against the design documents. Usually, a redlinedapproved for construction (AFC) P&ID will be generated andused as documentation of the completeness of the installation.The walk down should also yield a final punch list of incom-plete items.

Design and Construction of Facilities 163

Page 179: Active Pharmaceutical Ingredients

An example of typical items that would be verified in theIQ phase of validation and in the commissioning of theremaining facility:

� verify that the installation of the equipment complieswith the design specifications;

� verify that all required equipment, piping, electricaland instruments are installed;

� redlined AFC P&IDs are completed;� vendor documentation is available for all equipment;� full loop checks are completed;� all necessary utilities are connected and ready for

operation.

Any changes from the original P&IDs that are noted dur-ing the redlining function of IQ must be documented througha change control procedure, which is detailed in the validationplan. These changes, if approved and accepted, must then beincorporated into the final ‘‘as-builts’’ of the facility. It is cri-tical that the start-up team, maintenance, and the operationshave complete accurate documentation of the final as-builtconfiguration of the facility and the process. At the completionof the construction, the construction team for maintenanceand operations, and the start-up team must assemble a ‘‘turn-over package’’. This package will have at a minimum:

� as-builts drawings of the entire facility;� preventive maintenance information and require-

ments on all equipment;� suggested spare parts for all equipment, including

delivery time and pricing;� vendor manuals on all installed equipment.

The start-up of an API facility is a complex operationthat requires early planning and a complete understandingof all the components of the facility. An operations manualwith spare part and regular maintenance procedures mustbe in place on site for every component in the facility. A listof spare parts required for start up should also be assembledand these materials stored on site for use as required during

164 Mongiardo and Bobrow

Page 180: Active Pharmaceutical Ingredients

the start up of the facility. The most critical step in developinga complete start-up plan is to develop standard operating pro-cedures for the facility (SOPs). A complex API process facilitywill require SOPs be developed on every aspect of the opera-tion and including the maintenance procedures for thefacility. The SOPs are ultimately the responsibility of theoperations team for the facility, but may be developed by anengineering firm or consultant who is initially more familiarwith the specific details of the facility and the equipment.An important SOP at this phase of the project is the procedurefor the start-up and methodology for shut down procedures ofthe process. These procedures must show the order in whichthe equipment should be started and stopped, the setting ofvalve dampers, instruments and controllers to avoid damageto any equipment. For the initial start-up, which is normallydone with ‘‘water batching,’’ it may be necessary to compen-sate for the difference in the specific gravity of water to theprocess when the facility is in full operation.

XVIII. CONCLUSIONS

� The chapter outlines the steps required to design andconstruct a new API facility

� The business case� Process development� Design� Execution strategy and planning� Procurement strategies� Construction management� validation, start-up, and project turnover

In the review of the API facilities, distinct differencesbetween the requirements and those of a fine chemical facilityexist. The product is manufactured under cGMP guidelines.There are validation requirements for the facility, whichdocument how the facility was constructed, how the facilityand process will be operated, and how the facility will bemaintained. In most new product introductions, the primarydriver for API projects is the speed to market. The products

Design and Construction of Facilities 165

Page 181: Active Pharmaceutical Ingredients

have critical marketing considerations and schedule is ofparamount importance, consequently at the risk of highercost to construct. Because of the differences in the require-ments for API facilities vs. fine chemical facilities, both inthe construction and final operation, the initial planningstrategy for the project is critical to the success of the project.

The design and construction professionals must beknowledgeable in the specific cGMP requirements for APIfacilities. The construction personnel must plan for the properdocumentation of the facility throughout the constructionprocess so that the final facility can be validated and ulti-mately certified to manufacture product. The design and con-struction of an API manufacturing facility is a largeinvestment of time, resources, and capital. The proper plan-ning up front and the diligent effort to evaluate the economicoptions and interface these options with an overall projectschedule will produce a facility that operates as intendedand returns the predicted profit on the investment.

REFERENCES

1. ICH Q7A, International Conference on Harmonization. GoodManufacturing Guide for Active Pharmaceutical Ingredients.Recommended for Adoption at Step 4 of the ICH Process onNovember 10, 2000.

2. United States Pharmacopoeia (USP) 23 General Information.Water for Pharmaceutical Purposes. Chapter 1231. Rockville,MD: US Pharmacopoeia Convention, 1995.

3. 21 Code of Federal Regulations (CFR). Current Good Manufac-turing Practices for Finished Pharmaceuticals. Part 211.

166 Mongiardo and Bobrow

Page 182: Active Pharmaceutical Ingredients

5

Regulatory AffairsJOHN CURRAN

Merck & Co., Inc., Whitehouse Station, New Jersey, U.S.A.

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . 167II. Requirements for Submission of

Regulatory CMC Documents . . . . . . . . . . . . . . . . 169III. Contents of Regulatory Submissions—API Sections . . . 173IV. Registration Samples . . . . . . . . . . . . . . . . . . . . . 191V. The Review and Approval Process . . . . . . . . . . . . . 192VI. Preapproval Inspections . . . . . . . . . . . . . . . . . . . 195VII. Postapproval Change Evaluations . . . . . . . . . . . . . 196VIII. The Future . . . . . . . . . . . . . . . . . . . . . . . . . . . 199IX. Helpful References . . . . . . . . . . . . . . . . . . . . . . 200

I. INTRODUCTION

As we’ve entered the 21st century, regulatory agencies inthe United States (Food and Drug Administration, FDA),Europe, and around the world are placing an increasedemphasis on the manufacture and control of the active phar-maceutical ingredient (API), or as it has historically beenreferred to, the bulk drug substance. With the growingrequirements for chemistry, manufacturing, and control(CMC) documentation to support an original marketingapplication, as well as the advent of agency preapproval fieldinspections, virtually all aspects of the development andscale-up of the API are subject to regulatory review. Addi-tionally, stringent regulations exist, covering changes to

167

Page 183: Active Pharmaceutical Ingredients

the manufacture and control of the API following approval ofthe marketing applications, and field inspections arefrequently carried out by agencies to ensure ongoing compli-ance to the marketing application and to current goodmanufacturing practices (cGMPs).

Given the extent to which the current regulatory require-ments govern development, registration and maintenance ofCMC information for the API, the preparation of accurateand complete documentation has become more critical. It isincreasingly necessary for the regulatory area responsible forCMC within a company to develop the expertise needed to suc-cessfully register and maintain appropriate information anddocumentation on the API, and to assure that changing regula-tions are tracked, understood, and properly implemented. Thischapter is intended as an overview of current API regulationsas they exist in early 2004, and is designed to guide and assistthe CMC scientist in developing such expertise. While it is notthe intent to focus only on the regulations published by theU.S. FDA, it is clear that in most cases, FDA requirementsfor API are the strictest and most comprehensive. Hence, satis-fying FDA requirements often ensures that sufficient informa-tion and data exist to satisfy any worldwide regulatory agency.

This chapter presents an overall summary as a ‘‘snapshotin time’’ for an ever-evolving arena in the pharmaceuticalindustry, and the information contained herein is meant to sup-plement, not replace, the many excellent guidance documentspublished and maintained by regulatory authorities, world-wide, as well as the comprehensive documents published bythe International Conference on Harmonization (ICH).

The primary focus of this chapter will be on conventional,low-molecular-weight APIs manufactured by chemical syn-thesis. It is recognized that APIs also are derived fromfermentation, proteins, peptides, etc. Specific regulationsand guidelines exist for these compounds, which will not becovered in this work. This chapter will also focus mainly onthe regulations as they apply to innovator companies seekingapproval for new chemical entities. Separate but similarrequirements exist for the development and maintenanceof drug master files (DMFs) submitted by bulk chemical

168 Curran

Page 184: Active Pharmaceutical Ingredients

manufacturers that supply APIs to the industry (note—DMFsfor APIs are generally accepted in the United States, Canada,and the EU; other countries have varying requirementsregarding DMFs).

Please be aware the existing API regulations andguidelines published by the U.S. FDA and other regulatoryagencies are not comprehensive, and are often subject tointerpretation by the company as well as the regulatoryreviewer. It is therefore important to focus on sound scientificreasoning, supported by analytically valid data, in thepreparation of original and supplemental regulatory filings.The ability to clearly communicate the science and supportingdata can be a significant challenge for the CMC specialist.

II. REQUIREMENTS FOR SUBMISSION OFREGULATORY CMC DOCUMENTS

A. Investigational Compounds

An increasing number of worldwide regulatory authoritiesrequire the submission of relevant information supportingproposed clinical trials prior to the introduction of an experi-mental compound into man in their country. The terminologyused for these investigational submissions differs from agencyto agency. In the United States, the documentation is calledan Investigational New Drug Application (IND); in the U.K.its a Clinical Studies Exemption (CTX); in other Europeanand international markets, the term used is Clinical StudiesAuthorization (CSA); and in Canada the document is referredto as an Investigational New Drug Submission (INDS). Gen-erally, the document covers a specific clinical program for adesired therapeutic indication in a target patient population,and must be kept current throughout the clinical develop-ment program. Modifications of the indication or targetpopulation often require a separate original investigationalapplication.

The original investigational applications and subsequentupdates are formally reviewed by the agencies. For originalapplications, the clinical studies may typically be initiated

Regulatory Affairs 169

Page 185: Active Pharmaceutical Ingredients

after a prescribed time following submission, unless thecompany is informed otherwise by a particular agency (e.g.,clinical hold). The agency reviewers may and often do submitquestions to the company based on the investigational applica-tion, and should clearly communicate whether studies mayproceed prior to the resolution of the issues. Frequently,guidance for the ongoing development program is providedby the agency through these questions. Formal responses onall issues should be provided to the agency in an expeditiousmanner. Often, the responses include commitments for addi-tional investigations as development progresses.

This investigational documentation typically includesCMC information on the chemical characteristics, manufac-ture, control and stability of the API, and any formulationsplanned for evaluation in the clinic. For early developmentcandidates, often a brief overview of the API synthesis andsummaries of the characterization of the compound andapplicable specifications (tests and acceptance limits) are suf-ficient to allow initiation of clinical trials. As the developmentprogram progresses and the compound is to be introduced intolarger numbers of patients, more detailed supporting docu-mentation is generally required for the API. Significantchanges in the manufacture, characteristics, or controls forthe API must be communicated to the regulatory agenciesprior to use of the material in ongoing clinical studies,through updates or amendments to the investigational appli-cation. Periodic updates documenting other, less criticalchanges, should also be submitted during the clinical program(e.g., on an annual basis). Often, the updates to the investiga-tional filings provide useful references for generating histori-cal background information on the development program forinclusion in original marketing applications.

B. Application for Marketing

Research-oriented companies will evaluate a number ofinvestigational compounds for potential therapeutic indica-tions. A majority of the potential candidates do not survivethe safety and efficacy studies conducted as part of the clinical

170 Curran

Page 186: Active Pharmaceutical Ingredients

development program. Those that are found to be safe andeffective toward a specific indication must be registered withregulatory agencies worldwide, prior to their being made avail-able for sale in that market. The process by which compoundsare submitted for approval to market is very similar through-out most of the world. Specifically, a detailed application mustbe submitted to the recognized regulatory authority in thecountry, and that authority reviews the contents and renderstheir acceptance, conditional acceptance, or rejection of theapplication. As is the case for the investigational application,several different names are used to describe the marketingapplication in various countries. These include an NDA inthe United States, an NDS in Canada, and a PharmaceuticalDossier in Europe.

While the registration procedure is similar and therecent adoption of the common technical document (CTD)has begun to standardize the format for regulatory submis-sions, the content of the quality (CMC) section of the market-ing application required by different countries, particularly asit relates to information on the API, varies significantly. In anumber of countries, very limited, if any, information isrequired on the manufacture and control of the API, whilein others (e.g., the United States, the EU, Canada, Israel,and Australia) very detailed information and supporting dataare required on the characterization, manufacture, control,and stability of the API. A subsequent portion of this chaptercovers the information supporting the API to be included inmarketing applications for these concerned markets.

Following submission of the application, certain agencieswill perform a high-level review of its content to assure thatall basic elements are contained in the submission. Once theapplication is considered accepted for filing, the reviewingchemist or authority from each agency will perform a verydetailed evaluation of the CMC documentation, and whereappropriate will provide specific questions or comments onthe content of the documentation. The CMC questions oftenseek clarification or additional information or data on specificitems, or state concerns the reviewer has with the content orconclusions provided for certain aspects of the application.

Regulatory Affairs 171

Page 187: Active Pharmaceutical Ingredients

Formal responses to each question must be provided to theagency. Timing for responses varies from country to country,but generally the rapid submission of complete responses isdesirable to both the reviewer and the company. In mostinstances, delays in responding to the questions will result ina delay in the approval timing for the marketing application.It should be noted that knowledge of, and adherence to theexpectations documented in published agency guidelines gen-erally could minimize the number and=or severity of chemistryquestions received on a specific application.

Once the concerns on all aspects of the application,including CMC, are addressed to the satisfaction of thereviewing authority, an official ‘‘action letter’’ is typicallyprovided to the sponsor of the application, to formally allowmarketing of the product in that country. Sometimes, condi-tions for approval are stated in the letter. These conditionsshould be specific with respect to their impact on the market-ing of the product, and often must be satisfied before productis sold.

In the mid to later part of the 1990s, procedures wereestablished to allow for more timely review and approval ofmarketing applications in the both European Union and theUnited States. The review process to be used and the timingfor approval are defined by the local regulations, and aredependent upon the immediate therapeutic need for the pro-duct. The EU mutual recognition and centralized proceduresand the U.S. Prescription Drug User Fee Act (PDUFA) will bediscussed in more detail later in this chapter.

C. Postapproval Requirements

Following approval of the marketing applications, it is neces-sary that the CMC information on file with each regulatoryagency remains current and accurate. Unfortunately, thereare a wide variety of mechanisms that must be followed inthe various countries=regions to communicate changes thatare required or desired post approval. The mechanisms tobe used are often linked to the nature of the proposed change,and its potential to impact the quality (chemical and physical)

172 Curran

Page 188: Active Pharmaceutical Ingredients

or safety of the API and ultimately, the quality, potency,safety, or efficacy of the final drug product. Changes havinga moderate to significant chance of impacting any of thesecharacteristics generally require approval by the agency priorto the marketing of product containing API made or tested bythe changed route. In certain markets, changes having aminimal chance of impacting these characteristics can gener-ally be implemented (i.e., product using API made=tested bythe changed route can be marketed), and the agencies aresimultaneously or even subsequently notified of thesechanges via an appropriately defined mechanism. Furtherdetails on evaluating and reporting postapproval changesare provided later in this chapter.

Of critical importance in the maintenance of registeredinformation is the existence and implementation of strongchange control procedures. For the API, procedures shouldbe in place to address changes to the manufacturing process,(controls and parameters), specifications (analytical testprocedures and acceptance criteria), equipment cleaning pro-cedures, raw materials, and=or their acceptance criteria,packaging, etc. These procedures should be consistent withcurrent cGMPs and are often the focus of agency inspections.Defined change control procedures should also be included aspart of supply agreements with certain vendors (e.g., suppli-ers of key starting raw materials and packaging components),since changes made by these suppliers could result in theneed for regulatory submissions by the user, which poten-tially could require prior agency approval.

III. CONTENTS OF REGULATORYSUBMISSIONS—API SECTIONS

A. Content of Investigational Applications

As previously mentioned, a number of regulatory agencies,worldwide, require information on the characteristics, manu-facture, control, and=or stability of any investigational APIintended for experimental use in man, prior to initiation ofclinical trials in their country. The information required is

Regulatory Affairs 173

Page 189: Active Pharmaceutical Ingredients

intended to provide the reviewer with a general backgroundand understanding of the compound to be investigated, andis considerably less detailed than that required as part ofapplications for formal marketing approval. Regardless ofthe intended reviewing agency or the stage of developmentfor the compound, the main purpose of the investigationalapplication is to demonstrate that the API to be introducedinto man is adequately safe and is properly controlled. Theexact information required in an investigational applicationvaries from country to country, but generally consists of allor some of the items below:

� General information on the compound� Description of key chemical and physical properties

and characteristics� Proof of chemical structure� List of manufacturers� Method of manufacture (minimally a process flow

diagram)� Specifications (methods and acceptance criteria) for

the finished API� Discussion of impurities and degradation products� Analytical test results� Information on the analytical reference standard� Description of the container=closure system� Stability data

The level of detail required in the above sections can varysignificantly from country to country, and based on the stageof development for the compound. As the development pro-gram progresses, more detailed information is generallyincluded in the investigational applications, particularly forthe more sophisticated countries. It should be noted thatinvestigational applications may be submitted to a numberof different countries at different stages of the same develop-ment program, to support individual clinical trials to beconducted in that country.

For certain countries, there exist published guidancedocuments from the regulatory authority describing theexpected contents of the investigational application. This

174 Curran

Page 190: Active Pharmaceutical Ingredients

guidance should generally be followed when preparing anapplication, with any significant deviation from the guidancedescribed and justified. For some sections of the application,the level of detail for information to be provided is not well spe-cified in the agency regulations or guidelines; rather,experience with the respective reviewing agencies should dic-tate the detail provided. The typical contents of the sections,which comprise an investigational application, follow:

General information—This section should contain thefull chemical name, established in accordance with a recog-nized nomenclature system (e.g., IUPAC); the molecularformula and molecular weight; the stereo-specific chemicalstructure of the API; plus any internal codes used to designatethis compound within the document.

Description of key chemical and physical properties=characteristics—A physical description of the compound(color, form, and appearance) should be stated. A discussionshould be provided on existence of polymorphic forms, sol-vates or hydrates of the molecule supported by appropriatedata (e.g., thermal analyses or x-ray powder diffractiontesting). Available solubility data, specific (optical) rotationvalues for chiral compounds, the partition (distribution) coef-ficient, acid=base dissociation constants, pH, and hygroscopi-city data are generally also included for the selected form ofthe API.

Proof of chemical structure—Evidence supporting thestructural assignment for the API are provided, typicallyincluding appropriate spectroscopic or spectrophotometricevaluations and interpretations. At the investigational stage,direct proof of stereochemical conformation may be provided,but generally is not required.

List of manufacturers—The complete names andaddresses are provided for all sites that have or will beinvolved in the manufacture and testing of API for use duringthe development program. Typically, the names of suppliers ofstarting materials (i.e., compounds which impart a significantportion of the structure of the final API) as well as auxiliaryraw materials used in the synthesis are not given.

Regulatory Affairs 175

Page 191: Active Pharmaceutical Ingredients

Method of manufacture—The contents of this section willvary greatly depending upon the stage of the developmentprogram, and the intended country to receive the application.When a compound is being prepared for early clinical trials,the process steps are generally not yet well defined and arefrequently being modified or optimized. Thus, for original orearly-stage investigational applications, even in the some ofthe more sophisticated countries, it is suitable to submit aflow diagram of the chemical synthesis, possibly accompaniedby a brief, qualitative, narrative discussion of the processsteps. Initially, little detail is required for controls on thestarting materials and raw materials used in the synthesis.Only major changes to the route of synthesis should impactthe filed information early in the development program,requiring an amended investigational application prior touse of the material in the clinic.

As the development program progresses and the synth-esis becomes better defined, additional detail is typicallyprovided for the process description. This may occur aboutthe time in development when the synthesis is being scaledfrom laboratory to pilot plant scale equipment. At this time,acceptance criteria should be considered for key process rawmaterials, particularly the starting materials.

While the practices of individual companies differ, theprocess description provided for the manufacture of APIfor use in the preparation of drug product formulations forpivotal clinical (e.g., the definitive bioavailability study) orstability trials should be documented to a level of detailapproaching that to be included in the eventual marketingapplication. This will facilitate comparison of the synthesisof these key developmental batches with that to be used atthe final manufacturing site supplying API for marketed pro-duct. At this time, it may be practical to include in-processacceptance criteria for key reactions or intermediates, andacceptance criteria for all raw materials used in thissynthesis.

Specifications for the API—As is the case with thechemical synthesis, the analytical controls used to monitorthe identity, quality, and purity of the API also evolve during

176 Curran

Page 192: Active Pharmaceutical Ingredients

the development program. Hence, there is less of a regulatoryemphasis placed on those controls used in the early stages ofdevelopment, compared to those used in the release of mate-rial for pivotal clinical=stability trials late in the developmentprogram, and eventually those used for the release of API foruse in marketed product. Early in development, there isusually a very limited database to establish meaningfulacceptance criteria. Thus, the original investigational applica-tion may contain fairly broad or even tentative acceptancecriteria and brief narrative descriptions of the analyticalmethods that make up the early API specifications for thecompound. Specifications are generally included for physicalappearance, assay, impurity profile, water, final step solvents,inorganic impurities, and identity. Specific (optical) rotationshould be considered for chiral compounds, and a particle sizecontrol may be applied for certain APIs based on the intendedformulation use and their physical or chemical properties(e.g., those with low aqueous solubility). The initial assay pro-cedure may be nonspecific (e.g., titrimetric), until a suitablereference standard is prepared and qualified. Generally,formal method validation data need not be included in theoriginal application, although certain country requirementsand sponsor company practices may differ.

By the time API is being manufactured for pivotal clini-cal=stability studies, the analytical controls and the chemicalsynthesis are generally well established and a larger databaseexists for the compound. A specific assay procedure wouldhave typically been developed, and the impurity profile proce-dure would have been optimized to separate and quantitateimpurities and degradation products expected from the estab-lished route of synthesis. For compounds with limited numberof chiral centers, a chromatographic chiral purity proceduremay be applicable, replacing the specific rotation control.The need for control of physical properties relevant to thedrug product formulation should also be established, whereappropriate. Acceptance criteria can be modified to reflectboth process capabilities and the enhanced knowledge ofthe safety qualification limits for the investigational API.These better-defined tests and acceptance criteria should be

Regulatory Affairs 177

Page 193: Active Pharmaceutical Ingredients

included in the investigational application, through appropri-ate amendments, where necessary. In addition, validationdata for the analytical test methods may also be expected bystage in the program. The validation data should generallyfollow the guidelines provided in the general chapters of anapplicable compendium (e.g., United States Pharmacopoeia,European Pharmacopoeia, or Japanese Pharmacopoeia) orin the published ICH guidelines (Q2A=Q2B).

Discussion of impurities and degradation products—Thissection should include a discussion of impurities, which havebeen observed in development batches of the API, and to a les-ser extent those, which are potentially formed through theroute of synthesis. The focus of the discussion is generallyaround reaction byproducts, but should also include keyreagents and solvents (e.g., metals, final step solvents), aswell as known and potential degradation products of theAPI. The discussion should be based on the experiences fromthe batches made up to the time of submission, as tested bythe current analytical procedures. The qualification level ofthe impurities as determined through safety or other appro-priate studies should be addressed. As with the previoustwo sections, the understanding of impurities for an investi-gational compound is expected to evolve during development.Hence, early impurity discussions may be less detailed, andmay involve peaks for observed impurities, which are notyet structurally identified.

Analytical test results—This section should include testresults for batches of the investigational API manufacturedby the route of synthesis and tested against the acceptancecriteria contained in the investigational application. It is thechoice of the CMC specialist based on company practiceswhether a comprehensive tabulation of data for all clinicalAPI batches should be provided. In all cases, pertinent infor-mation such as batch size, site of manufacture, date of manu-facture and clinical use should be provided for each batch.

Information on the analytical reference standard—Once asuitable reference standard for the API is established, appro-priate information on its manufacture and results from char-acterization studies should be included in the investigational

178 Curran

Page 194: Active Pharmaceutical Ingredients

application. Often it is possible to refer to the synthetic routein the application for the preparation of the reference stan-dard, and to add any additional purification steps used. Thissection should also contain information on other requiredreference materials (e.g., an impurity standard).

Description of the container=closure system—A briefdescription of the primary and minimally functional second-ary materials used in the packaging of the finished develop-mental API should be provided (e.g., double high-densitypolyethylene liner inside a fiberboard container=closure).Consideration should also be given to including appropriateacceptance criteria for the primary (i.e., product contact)material. The container=closure described in this sectionshould be consistent with that used for stability testing ofthe compound, or exceptions should be noted.

Stability data—Data from stability studies conducted onthe investigational API should be presented, along with thetest methods used for the studies, and the conclusionsreached from these studies (including the recommended sto-rage temperature and conditions). Stability studies carriedout early in development may be conducted against internalprotocols, and may focus on establishing suitable storage tem-peratures and=or container=closure systems. Typically, for-mal studies for long-term and accelerated stability areconducted on material made for use in pivotal clinical and=orformal drug product stability trials later in the developmentprogram. Where practical, these studies should be conductedin a manner consistent with applicable guidelines publishedby the International Conference on Harmonization (ICHTopic Q1A). This will allow data from these stability studiesto be used to support preparation of the eventual marketingapplication. (Note: ICH Topic Q1A does not specifically coverstability studies to support clinical trials, but does cover stu-dies to support marketing applications.) Results from stressstability studies on the API (e.g., light, oxidative, acid, base,thermal, solution) should also be reported.

As the program progresses, certain agencies now expectthat details of the stability protocol used for testing of theAPI be included in the investigational application.

Regulatory Affairs 179

Page 195: Active Pharmaceutical Ingredients

B. Content of Marketing Applications

The marketing application is the mechanism by which theregulatory reviewing agencies in most countries will grantpermission to sell the subject product in their market. Thegoals for a well-prepared API section of the application areto convince the reviewer that the compound:

� is well characterized;� is manufactured at production scale using a rugged

and well-controlled synthesis, which consistently pro-vides material of comparable or better quality thanthat used in the development program;

� is tested using validated analytical procedures whichshow that each batch meets meaningful, justifiedquality requirements (acceptance criteria) reflectingthe quality of the clinical=safety material and thecapabilities of the manufacturing process.

Further, the marketing application demonstrates thatthe API is adequately stable, when stored under defined con-ditions, to assure that its quality is maintained at least untilit is used in the manufacture of the formulated drug productto be marketed.

Historically, there has been a wide range of informationon the API expected by worldwide regulatory agencies forinclusion in marketing applications. With the recent publica-tion of guidance on the common technical document (ICHM4Q), the format and high-level content of API informationrequired for a marketing application has been established.While ICH has been developed by and for the United States,EU, and Japan, other regulatory agencies are also followingthis guidance for new drug applications. Under the CTD,the application should include the following API sections:

� General information

NomenclatureStructureGeneral properties

� Manufacturing information

180 Curran

Page 196: Active Pharmaceutical Ingredients

Sites of manufactureDescription of the manufacturing process and pro-

cess controlsControls on starting raw materialsControls on critical steps and intermediatesProcess validation or evaluationManufacturing process development

� Characterization of the API

Elucidation of the chemical structureDiscussion of impurities

� Specifications for the finished API

Acceptance criteriaTest methodsAnalytical validation dataBatch analysis resultsJustification of the recommended specifications

� Reference standard information� Container=closure information� Stability

Summary and conclusionsPostapproval stability commitmentsStability data

While the format and high-level content of the applicationhas been established by ICH, the expectations for the exactcontent (e.g., the type of information and the level of detailrequired) in the above sections still differ for different mar-kets. Generally, the contents of a marketing application arean extension of the information in the investigational applica-tion, with increased levels of detail in certain sections, andclearly defined ‘‘bridges,’’ where appropriate, between thematerial studied in development and that to be routinelymanufactured for market. Thus, properly prepared and main-tained investigational applications will serve as a good startingpoint for the preparation of the marketing application for acompound. For APIs purchased from a contract manufacturer,

Regulatory Affairs 181

Page 197: Active Pharmaceutical Ingredients

a significant portion of this information may be providedthrough reference to a DMF, where the regulations allow(e.g., United States, EU, Canada). The typical content for theAPI sections of the marketing application (or DMF) arewell defined in various guidance published by the respectiveagencies or by ICH. A brief overview of the type of informationprovided in an API section of a marketing application follows.

1. General Information

Nomenclature—The recommended international nonproprie-tary name (INN), U.S.-adopted name (USAN) and othernational names are included, if available, along with the fullchemical name(s) and any internal code names or numbersfor the API. Additionally, trade names for the intended formu-lation(s) and the CAS number for the API may be provided, ifestablished.

Structure—This section will generally include a full,stereo-specific chemical structure for the API, along with itsmolecular formula and molecular weight (relative molecularmass). If the compound is a salt or solvate, the molecularweight of the core molecule should also be provided.

General properties—The information provided for aspecific compound may differ based on the exact molecule,but usually includes the following information: physicalappearance, thermal behavior, solubility, chirality and specific(optical) rotation, crystallinity and polymorphism, hygroscopi-city, partition coefficients, solution pH, and acid=base dissocia-tion constants. The majority of this information is generatedduring the early development of the compound, and includedin the investigational application as discussed above. Undercertain circumstances, it may be necessary to evaluate theproperties of large-scale pilot batches or even early productionscale batches to demonstrate consistency with the propertiesreported for the early development batches. Any differencesin these properties should be discussed as part of the market-ing application, and sufficient evidence should be provided todemonstrate the equivalence of the pilot=production materialto material used during safety and clinical trials.

182 Curran

Page 198: Active Pharmaceutical Ingredients

2. Manufacturing Information

Sites of manufacture—All sites involved in the manufactureand testing of the finished API and=or key process intermedi-ates for commercial purposes are listed in the application sub-mitted to the U.S. FDA and certain other markets, along withtheir complete addresses (street addresses are generallyrequired as opposed to P.O. Boxes). Other countries=regionsfocus only on the manufacturing site of the finished API,while in others this information is not submitted at all. Inthe U.S. NDA, the sites listed in the application, includingcontract manufacturing and testing sites, are potential candi-dates for agency inspection during the approval process.Thus, the sites should be ready to manufacture or test thespecified API or intermediate at the time of filing (e.g., theprocess equipment and=or supporting documents are inplace).

If the entire API and=or key process intermediates aremanufactured at contract facilities, the information requiredin this section differs from country to country. In the UnitedStates, the contract manufacturer for an API or intermediateis generally identified in the filing, along with specific refer-ence to their U.S. drug master file. A letter from the contractmanufacturer, allowing FDA review of the DMF as part of thesponsor marketing application, is also needed. In the EU atthis time, the regulations allow DMF reference only for thefinished API.

Manufacturing process description—Typically, this sec-tion contains a schematic flow diagram of the synthetic routeto be used to manufacture the API for commercial purposes,and a textual description of the processing steps. The levelof detail for the synthetic process that is required by agenciesdiffers significantly from country to country, and also in somecases from reviewer to reviewer. Certain countries require noinformation on the manufacturing process, some will acceptonly a flow diagram, while others expect sufficient informa-tion such that they can understand the manufacturing routeand are assured that suitable controls are in place to guaran-tee consistent quality batch to batch. The expectations also

Regulatory Affairs 183

Page 199: Active Pharmaceutical Ingredients

differ based on the type of synthetic process used (e.g., moredetail would be expected for fermentation processes than forstraightforward coupling reactions). The level of detail typi-cally provided by specific companies in their marketing appli-cations also varies greatly. The factors, which impact the levelof detail included in the process description, are many, andare often influenced by the philosophies of the individualcompany; hence, they will not be discussed in this work.

Controls on starting materials—Suitable analyticalcontrols (tests and acceptance criteria) should be included inthe application for all raw materials used in the filed manu-facturing process description, including those defined asstarting materials. The controls required for specific rawmaterials are dictated by the role the compounds play in thesynthetic process. Starting materials, which contributedirectly to the structure of the finished API, generally requirecontrols on identity and purity. Raw materials which impactor drive the quality of the API or an intermediate (e.g., achiral reducing agent) should have controls appropriate totheir specific role in the synthesis, while reagents and mostsolvents may require only identity testing to assure their suit-ability for use in the process. If the synthesis utilizes morestructurally complex starting materials, additional controlssuch as impurity limits and chiral purity testing may beappropriate. The acceptance criteria for raw materials shouldbe consistent with the demonstrated capabilities of the pro-cess. Evidence (experimental data) should exist that rawmaterials with the specified quality can be successfully pro-cessed forward. This evidence would not typically be includedin the original filing, but may be needed to respond to anagency question or may be reviewed during an inspection.

A brief synopsis of the test methods for raw materials isgenerally sufficient, and validation data have, to date, notbeen required in the original application. For items acceptedbased on supplier certificates, it is still necessary in mostcases to list either the company’s or the supplier’s tests andacceptance criteria.

It should be noted that currently there are significantdiscussions ongoing between the regulatory agencies and

184 Curran

Page 200: Active Pharmaceutical Ingredients

industry on the development of a suitable, mutually accepteddefinition of a starting material. Unfortunately, no such defi-nition appears imminent. Many companies elect to reviewtheir starting material designations with the regulatory agen-cies, particularly FDA, prior to submission of the marketingapplication.

Controls on critical steps=intermediates—Many regula-tory agencies expect that companies will monitor qualitythroughout the process. Thus, they would expect that in-process controls, including tests and acceptance criteriaapplied to critical steps (e.g., completeness of reaction) andkey (isolated) intermediates, be documented in the applica-tion. Practices and expectations defining critical process stepsand for inclusion of in-process controls in a regulatory processdescription vary significantly from company to company andfrom agency to agency. Additionally, in-process testing maybe used to demonstrate the removal of certain early stepimpurities (e.g., reagents or solvents) during further proces-sing, thereby eliminating the need for direct controls appliedto the finished API. Usually, in-process controls should trig-ger appropriate corrective actions if acceptance criteria arenot achieved. This can involve, for example, increasing thereaction time, charging additional raw materials, or reproces-sing an intermediate. A suitable corrective action should begiven for each in-process control provided in the synthesis.Typically, summaries rather than fully detailed in-processtest procedures are included in the application, and validationdata are not provided.

Recently, more emphasis is being placed on the use ofprocess analytics technology (PAT) for on-line or in-vesselreaction monitoring. The requirements for documenting andsupporting the use of PAT are presently being developed byindustry and the regulatory agencies.

Process validation=evaluation—Presentation of informa-tion in this section of the CTD is a new requirement, and isbeing actively defined by various agencies at this time. Therequirements may be different depending upon the nature ofthe API synthesis, and the end use of the API (e.g., formal pro-cess validation data may be required for the sterility operations

Regulatory Affairs 185

Page 201: Active Pharmaceutical Ingredients

for sterile APIs). Consultation of appropriate guidance docu-ments, when available, is therefore recommended.

Process development history—A brief discussion of thedevelopment of the synthetic process may be expected in themarketing applications. This section allows the company toestablish the bridge from the synthesis used to manufactureearly development safety samples, clinical materials, pivotalclinical=stability batches, and API for commercial product.This bridge is particularly valuable for reviewers in the moresophisticated markets who may not have previously reviewedthe processes contained in the investigational applications.Again, the level of detail provided for this section will varyconsiderably.

3. Characterization

Elucidation of the chemical structure—Data are required tosupport the structural assignment for the API made by theprocess described in the application. Typically, data are gener-ated using appropriate spectroscopic or spectrophotometrictechniques. Interpretations of the data are also included. Addi-tionally, results from a study (e.g., single crystal x-ray) provingthe stereochemical conformation of the molecule should be pro-vided. Recently, certain agencies have also been requestingevidence of the chemical structure of starting materials, pro-cess intermediates, and key synthetic impurities.

Discussion of impurities—The contents of this sectionshould be suitable to demonstrate to a reviewer that the com-pany understands the origins of, and has in place adequatecontrols for, impurities which may be present in the API madeby the filed process, with a focus on those impurities actuallypresent in batches during the development program. The dis-cussion should demonstrate that the controls in the applicationare consistent with levels of impurities qualified in safety stu-dies, and with appropriate ICH guidance (Q3A). The level ofdetail required to achieve these objectives varies dependingupon the complexity of the synthesis. As with the processdescription and process history discussions, the detail of thissection is often driven by internal company philosophies.

186 Curran

Page 202: Active Pharmaceutical Ingredients

Regardless of the level of detail, the discussion should includestructurally related impurities derived from the route ofsynthesis, process solvents, reagents and their byproducts(e.g., inorganic impurities), and potential degradation pro-ducts. For chiral compounds, potential isomers should alsobe addressed. Typically, the chemical structure and a briefdiscussion of the formation or origin of the impurity aregiven. The fate of the impurity in further processing is alsoincluded, along with the levels observed in actual batches.A discussion of the methods used to monitor these impuritiesmay be provided or referenced. For impurities that arenot directly controlled through the finished API specifica-tions, justification for omission of a direct control may beprovided in this section or in the justification of specificationssection.

4. Specifications

Acceptance criteria—A listing of the acceptance criteria to beused for release of the finished API is required. These limitsshould be developed according to established practices withinthe individual company, taking into account the demonstratedprocess capability (batch data) and safety=toxicity data for thecompound and its potential impurities. Guidance documentshave recently been published, which should be considered dur-ing the development of the acceptance criteria for the API.Specifically, the ICH has issued guidance documents Q3A—Impurities Testing Guideline: Impurities in New Drug Sub-stances and Q3C—Impurities: Residual Solvents.

Test methods—The analytical procedures used to test thefinished API should be provided in the application. The levelof detail is again subject to company philosophy; however, suf-ficient detail should be included to provide the reviewer witha solid understanding of the method. In certain countries, thetest methods may actually be run in an agency or contractlaboratory, to confirm results on samples provided with theapplication. Certain tests can be performed using establishedcompendial methods, with the compendial method referencedin the application. Often it is helpful to attach copies of these

Regulatory Affairs 187

Page 203: Active Pharmaceutical Ingredients

compendial methods to the application to facilitate review,particularly in countries, which do not follow the referencedcompendia.

Analytical validation data—Complete validation dataare required showing the analytical methods to be suitableand rugged for the control of the API. Several guidance areavailable on analytical method validation, including UnitedStates Pharmacopoeia (USP) General Chapter <1225> , Vali-dation of Compendial Methods, and the ICH guidance docu-ments Q2A—Validation of Analytical Methods: Definitionsand Terminology and Q2B—Note for Guidance on Validationof Analytical Procedures: Methodology. Following these orsimilar guidance documents should assure suitable validationdata in the regulatory application. For tests which referenceestablished compendial methods, revalidation of the proce-dure is generally not required; however, it may be necessaryto demonstrate that the method can be suitably applied tothe subject API.

Batch analysis results—Complete batch analysis datashould be included for key batches prepared throughout thedevelopment program, including, if practical, material manu-factured at the site(s) that will supply material for commer-cial purposes. These test results should be provided againstthe specifications in place at the time of the release of thebatch for its intended use. Often, it is valuable to include ret-rospective analyses of key batches using techniques estab-lished or modified subsequent to their initial release. Thiscould be particularly beneficial for impurities, assuming theimpurity profile method(s) were modified during develop-ment. It is also expected that information on the manufacture(date, size, location) and use of each listed batch (clinical,safety, stability, market product, etc.) be included in theapplication.

Justification of the recommended specifications—It isoften helpful to present the rationale used for establishingthe acceptance criteria proposed for the API in the marketingapplication. This will provide the reviewers with an under-standing of the thought processes used to establish the controls(e.g., are impurity limits based on safety=toxicity studies,

188 Curran

Page 204: Active Pharmaceutical Ingredients

analytical results, or both? was ICH guidance followed, whichbatches were used to set acceptance criteria? were limits deter-mined statistically? etc.). Also, this section can be utilized tosupport the decision to omit certain direct controls on theAPI. Providing this information does not assure acceptance ofthe proposed specifications by all reviewers; however, it mayhelp to focus reviewer comments related to the recommendedacceptance criteria.

5. Reference Standard Information

Method of preparation—The preparation of the primary refer-ence standard for the API should be provided, with a focus onthe means of purification. The synthesis used to manufacturethe batch can be summarized, or referenced to another sectionof the application, as appropriate.

Characterization data—Full characterization datashould be supplied for the primary reference standard, includ-ing the results from analytical testing and spectral character-ization. The assigned purity of the reference standard shouldbe clearly designated.

If more than one reference standard lot has been madeduring the development program, and subsequent lots arecharacterized against the original reference standard, itmay be appropriate to provide information on the manufac-ture and characterization of the original lot in the application.If the reference standard differs from the API (e.g., a morestable salt form or a solution is used), this should be indicatedand the rationale provided. Information should also be pro-vided for all other standard materials cited in the application,including, for example, those used to establish chromato-graphic system suitability and those used for analysis ofstarting materials, impurities, or intermediates.

6. Container=Closure Information

A description and statement of the composition of the container=closure system used to store and=or ship the finished APIshould be provided. Where a nonroutine system is used, adrawing is often very useful. Although the requirements

Regulatory Affairs 189

Page 205: Active Pharmaceutical Ingredients

differ, it may also be appropriate to discuss any controls inplace for acceptance, particularly of the primary (product con-tact) components, and possibly the cleaning of the containerclosure system.

7. Stability

Please note that ICH Q1A, Q1B, Q1C, Q1D, and Q1F provideexcellent and current guidance on stability testing thatshould be considered in the preparation of this section ofthe application.

Summary and conclusions—A summary should be pro-vided describing how the stability characteristics of the APIwere determined during the development program. The dis-cussion should include a review of the stability-indicating testmethods used in the studies, the batches tested, the storageconditions, and containers evaluated, and the final recom-mended storage conditions for production material. The useof bracketing or matrixing should be discussed and justified.A shelf life or retest period should be proposed based on ana-lysis of the available data at the time of submission. Testinggenerally includes both long-term and accelerated studies,using the conditions described in the ICH guidance documentQ1A—Stability Testing Guidelines: Stability Testing of NewDrug Substances and Products. Additionally, stress studiesexposing the compound to acid, base, high temperature, light,and oxidation, should be reported (these studies may be per-formed as part of the validation of the impurity profilemethod, to demonstrate selectivity as well as the stability-indicating nature of the method). Solution stability studiesmay also be performed and reported. The batches used toestablish shelf life or retest dating for the API should havebeen manufactured minimally at pilot scale using a synthesisequivalent to that to be used for preparation of material forcommercial purposes.

Postapproval stability commitments—Agencies expectthat companies will have in place a procedure for routinemonitoring of the stability characteristics of API productionmaterial. Recently, several agencies have required that the

190 Curran

Page 206: Active Pharmaceutical Ingredients

postapproval API stability protocol be provided in the marketingapplication. The nature of the commitment contained in theAPI stability protocol will vary based on the stability charac-teristics of the material and the practices of the individualsponsor company.

Stability data—Tabulations containing the actual testresults from the studies summarized in the stability sectionof the application should be provided. These results shouldsupport the conclusions stated in the document. Any devia-tions from the recommended acceptance criteria should benoted and explained. Data from stress studies can often bereferenced to the analytical methods validation section ofthe application, or vice versa.

C. Other Documentation Included in MarketingApplications

The CTD format allows other, regional specific information, tobe provided in a separate section of the application. Therequirements for this section are often described in detail inthe individual agency guidance documents. The CTD formatalso specifies sections for inclusion of references and attach-ments, as appropriate. Additionally, the CTD includes a qual-ity overall summary (QOS) of the more detailed informationprovided in the application. The contents of the QOS relatedto API are fully described in ICH M4Q, and represent a briefoverview of the main API section. The QOS may be used dur-ing the review of the application by other reviewing disci-plines (e.g., clinical, pharm=tox, etc.). The QOS in essencehas eliminated the need for a specific expert opinion reportand summary tables in the EU; however, it is expected thatthe contents of the document submitted to the EU have beenreviewed and accepted by an appropriate quality expert, andthat this expert is suitably identified in the application.

IV. REGISTRATION SAMPLES

Samples of the API, reference standards, and key impuritieswill be requested by certain agencies so that they can perform

Regulatory Affairs 191

Page 207: Active Pharmaceutical Ingredients

the test methods contained in the marketing application.These samples plus certificates of analysis, and sometimesthe analytical columns and reagents required for testing,are often sent to the reviewing agency or their designatedtesting laboratory shortly after submission of the marketingapplication. The exact quantities required are driven by thetest procedures; however, it is not uncommon for an agencyto request excessive quantities, particularly of authenticimpurity samples. In the United States, samples of the APIare also required for retention as forensic samples.

V. THE REVIEW AND APPROVAL PROCESS

The period during which the regulatory agencies are review-ing the contents of the marketing application can be verydynamic, visible, and sometimes intense. Since the goal ofthe company is to get the product to market in each countryin the fastest period of time possible, there will often be pres-sure for each individual discipline to reach agreement withtheir reviewers on the final content of their respective sec-tions of the application. This will often require negotiation,clarification, adding information, or providing more detail toaddress the concerns of the reviewer. Frequently, compro-mises must be reached. Occasionally, the company mustaccept undesired decisions by the reviewer in order to gainapproval. For the API section of the application, the criticaldiscussions often will focus on the specifications applied tothe finished API. Obviously, it is important that meaningfuland justifiable test methods and acceptance criteria areapproved, and that the manufacturing process is capable ofroutinely producing material which meets these controls.

A definite strategy should be established for developingresponses to agency reviewer questions. It is important thatresponses are well thought out, provide an answer to the spe-cific question, are based on sound scientific rationale anddata, and do not provide excessive additional information thatmay prompt further questions or concerns. It may be appro-priate to seek clarification of the question from the reviewer,

192 Curran

Page 208: Active Pharmaceutical Ingredients

particularly in cases where the questions are translated fromanother language.

While the path of least resistance during the approvalprocess could be to agree with the reviewer comments andrecommendations, this approach frequently does not bestserve the interest of the company. It is often well worth theeffort to defend the original recommendations or conclusionsin the application, or to seek a reasonable compromise posi-tion. It may be necessary to establish direct dialogue withthe reviewer, if possible, to avoid prolonged discussions on aproblematic issue.

Questions from regulatory agency reviewers are valuablelearning tools, and often aid the company in the preparationof future applications for a particular market or markets.These learnings should be communicated back to the develop-ment areas, as much of the content of the application has itsorigins in early development.

Finally, it is critical to keep appropriate impacted areas(e.g., manufacturing and testing sites, raw material purchas-ing areas, etc.) within and outside the company aware of theongoing discussions and the final outcome of the review andapproval process. Impacted areas should be directly involvedin the preparation, or at least review, or regulatory responsesand commitments.

It is well recognized by industry and regulatory agenciesalike that review and approval of the application has thepotential to be a long and difficult process. In the UnitedStates and the EU, regulations have been enacted which seekto limit the duration of the majority of regulatory reviews,provided the information contained in the application meetsminimum standards of completeness and acceptability.

The United States adopted a process known as thePrescription Drug User Fee Act (PDUFA), which defines theexpected length of time for review of an original application.In exchange for a fee, the FDA agrees to render a decision onthe application (approval, approvable under stated conditions,or not approved) within a period of time dictated by the natureof the new drug product. Currently, decisions on applicationsserving major therapeutic needs (1P compounds) will be

Regulatory Affairs 193

Page 209: Active Pharmaceutical Ingredients

targeted for 6 months. Other applications accepted underPDUFA should be reviewed within a 10-month period. Sincethe timeline is aggressive from an agency prospective, thereis more pressure on the sponsor to provide a complete and accu-rate original application, to be ready for preapproval inspec-tions (PAI) of facilities contained in the application, and torapidly achieve resolution of issues raised during the review.

In the EU, marketing applications must now be submit-ted for approval through one of two processes, the centralizedprocedure (CP) reserved for applications serving major thera-peutic needs, and the mutual recognition procedure (MRP),used for all other applications. Under both procedures, theoriginal application is initially reviewed by one of the EUmember states. Under the CP, the initial reviewing country(rapporteur) and a corapporteur are selected by Committeefor Proprietary Medicinal Products (CPMP) of the EuropeanAgency for the Evaluation of Medicinal Products (EMEA).The rapporteur performs an initial assessment of the applica-tion, and this assessment is then reviewed by the remainingEU member states. A scientific opinion is rendered on theapplication 210 days after submission. Questions on the appli-cation should be communicated to the sponsor by day 120 inthe process. Once approval is reached, the application isconsidered approved in all EU markets.

The mutual recognition procedure in the EU involves theapplicant selecting a reference member state (RMS) for initialreview of the application. This review process is less struc-tured in the EU guidelines, and timing may vary dependingon the selected RMS and the complexity of the application.The RMS will generally issue incomplete letters during theirreview, seeking additional information to support theirapproval of the application. Once the application is approvedby the RMS, the applicant generally will update their docu-mentation to reflect the outcome of the approval process(e.g., the CMC documents may require updating to reflectupdated or additional information or controls). At the sametime, the RMS prepares assessment reports to be shared withthe remaining EU member states. The application is thensubmitted for mutual recognition to some or all remaining

194 Curran

Page 210: Active Pharmaceutical Ingredients

EU concerned member states, which involves a defined90-day review period. Timing for comments=questions, com-pany responses, discussions, and final action are well definedand quite aggressive. Under MRP, responses to all issuesraised by the EU member states must be submitted within7 days of receipt of the questions, which occurs approximately2 months following initiation of the process. The final decisionon the application (approval, company withdrawal of theapplication in some or all member states, or binding arbitra-tion) is then made by day 90.

The exact policies governing the EU centralized andmutual recognition procedures and the U.S. PDUFA law arewell defined and published, and will not be reviewed here infurther detail.

VI. PREAPPROVAL INSPECTIONS

With the recent advent of regulatory agency site inspectionsduring the approval process for marketing applications, anincreased emphasis must be placed on the readiness of thesite to manufacture and test the specified API or processintermediate. In particular, compliance to current cGMPsand good laboratory practices (cGLPs) must be assured, andsupporting data must be available and in good order to sub-stantiate the information submitted in the application.Inspections may occur at any time following submission ofthe application, but usually will not be initiated until theapplication has been judged to be acceptable for review bythe agency. The exact timing for the inspection may benegotiated, and its duration will depend on the extent ofinformation to be covered, and the observations made.

Most companies will attempt to assure site readinessthrough internal audits of company and contract facilitiesprior to submission of the filing. Sites specified in the applica-tion should have manufactured the API or intermediate atthe time of filing, or at least be suitably equipped and pre-pared to do so. All process (e.g., master batch records) andcontrol (e.g., analytical quality standards) documentation

Regulatory Affairs 195

Page 211: Active Pharmaceutical Ingredients

should be completed and approved. Appropriate standardoperating procedures (SOPs), equipment qualification andcleaning documentation, and employee training recordsshould be in order and available for inspection.

The importance of a successful inspection cannot be over-stated, as significant concerns uncovered during a pre-approval inspection can delay and even prevent approval ofthe application. Deficiencies may also be considered indicativeof deeper problems within the company, and may have impactbeyond the subject filing.

VII. POSTAPPROVAL CHANGE EVALUATIONS

Interactions between the company and the regulatory agen-cies do not end with the approval of the marketing applica-tion. In fact, most agencies place as much emphasis onpostapproval activities as they do on approval of the originalfiling. The reason for this practice is simple; the agenciesare charged with assuring the drugs being marketed in theircountry are and remain safe and effective. Thus, changes tothe information approved in the marketing application mustbe reviewed with or at least communicate to the respectiveagencies, often before drug product containing API made ortested under the change can be placed on the market.

The content of a postapproval submission reporting achange to the manufacture or control of the API is dictatedby the requirements of the different regulatory agencies, asdocumented in published guidelines. In the late 1990s andinto the early 2000s, United States and EU agencies placeda significant emphasis on better defining the impact ofchanges related to the API, and in communicating the extentof supporting data that should be provided.

The EU published and later modified their guidelines forsubmission of variations to marketing applications. Theseguidelines list the type of changes that can be submitted asType I (minor) variations, and contain the necessary informa-tion and documentation required to support the proposedchange. A number of the defined Type I (minor) variations

196 Curran

Page 212: Active Pharmaceutical Ingredients

reflected changes impacting the API. Any change notspecifically defined as Type I in the EU guidelines, must befiled as a Type II (major) variation. Subsequently, the EUfurther defined minor changes that can be implemented assoon as documentation is provided to the agency (Type IA)and those, which require a brief waiting period to allow aninitial broad review before implementation (Type IB). TheType IA=IB designations technically apply to products regis-tered either by the Mutual Recognition or Centralized Proce-dures; however certain EU agencies are following these sameguidelines for older products registered nationally in theircountry.

Under the current mutual recognition and centralizedprocedures, variations must be reviewed with, and approvedby all concerned member states. The variation is providedunder mutual recognition through the reference memberstate for the original application, and defined timelines existfor review and approval of the variation, based on whetherit is a Type I or II submission. The timelines do not includethe time required by the application holder to respond toany questions raised by any of the concerned member states.

For variations submitted to the EU, it is necessary to uti-lize the same CTD format used for new applications, even ifthe original filing was prepared in an older, approved format(e.g., Part II). This requirement has met resistance from a por-tion of the industry, in that it implies the need for conversionof existing registration documents to CTD format. EuropeanUnion has not mandated such conversion; rather they havestrongly recommended that the conversion be performed.

In the United States, two guidance documents haverecently been published covering postapproval changes tothe API. In November 1999, FDA released their Guidancefor Industry—Changes to an Approved NDA or ANDA. Thisdocument covered API and drug product changes, focusingon the filing mechanism rather than the required supportingdata. The November 1999 guidance utilized the premise ofassessing change for the potential adverse impact it may haveon the safety, quality, or efficacy of the drug product. Changeswith significant potential for adverse effect would require

Regulatory Affairs 197

Page 213: Active Pharmaceutical Ingredients

formal agency approval before implementation (priorapproval supplement). Changes, which present a moderatedegree of potential adverse impact, could be implementedupon submission of the required documentation to FDA, or30 days after that submission [changes being effected (CBE)or CBE-30]. Those changes with minimum potential negativeimpact could be implemented and submitted in the annualreport, an update required by FDA for all active NDAs, basedon the anniversary date of initial U.S. approval. Food andDrug Administration published an update to this guidancedocument in April 2004.

In February 2001, the FDA issued the Guidance forIndustry—BACPAC I: Intermediates in Drug SubstanceSynthesis. This guidance provides filing mechanisms as wellas supporting data recommendations for all type changeswhich impact API manufacture and control prior to theformation of the agency defined ‘‘Final Intermediate’’ in theprocess. The BACPAC I guidance reenforces FDA’s use ofappropriate risk assessment in determining the filing require-ments for a change. Under BACPAC I, most changes can bereported either in the annual report or via CBE=CBE-30,since changes made early in the synthetic process, or changesto controls on materials early in the synthesis, are logicallyless likely to have significant potential for adverse impacton the drug product safety, efficacy, or quality. BACPAC Ialso introduced the concept of being able to evaluate theimpact of the change at an appropriate, well-controlled inter-mediate, which can eliminate the need to take change mate-rial forward to the final API to support the supplement.

The Food and Drug Administration is currently draftingfor industry review a follow-up guidance (BACPAC II) cover-ing steps from the defined ‘‘final intermediate’’ through thefinished API. In general, it is expected that changes madeat this late stage will require more regulatory review. Hence,many changes covered under BACPAC II are expected toresult in the need for prior approval supplements.

For all postapproval supplements=variations worldwide,the key for the company is to provide evidence that thechange does not impact the quality (e.g., impurity profile,

198 Curran

Page 214: Active Pharmaceutical Ingredients

physical properties) of the API. It is often sufficient todemonstrate comparability of pre- and postchange material,provided adequate analytical procedures exist to do so. Wherethis comparability is not achieved, it is necessary to show con-clusively that the change does not impact drug product safety,quality, or efficacy, based on either performance tests on theresulting drug product or even by repeat of trials to show bio-equivalence to prechange material.

One key factor that dictates often timing for filing ofchanges to the API synthesis in a number of markets is theneed to provide comparability data on material manufacturedat full scale, or at least at pilot scale. Since drug product madewith the process change material often cannot be released forsale to a market until approval of the change is obtained, thepreparation of full scale batches coupled with prolongedapproval processes tend to have negative business implica-tions for a company. Thus, changes requiring prior agencyapproval are generally undertaken only for need or significantlong-term benefits (e.g., financial savings).

A tool recently introduced in the EU to facilitate moreefficient postapproval change control is the certificate of suit-ability (CoS). Obtaining a CoS involves the review andapproval by the European Pharmacopoeial Commission ofthe current API characterization, manufacturing and controldata for an already registered API. The commission thengrants the CoS, which can be referenced in existing market-ing applications, replacing the API section of that application.The CoS must be maintained, and therefore changes made tothe API must be filed to the CoS. At this time, the EU onlyrecommends the use of a CoS for established APIs; however,legislation has been proposed that would mandate thisapproach for APIs that have monographs in the EuropeanPharmacopoeia.

VIII. THE FUTURE

It is hopefully evident from the above discussions that regula-tions governing the API are continuing to evolve. At this time,

Regulatory Affairs 199

Page 215: Active Pharmaceutical Ingredients

the requirements still differ from country to country, andthese differences have and continue to place an undue burdenon the industry. The International Conference on Harmoniza-tion has taken an important initiative in developing and pub-lishing meaningful guidance documents for industry, and thepositive impact of their activities is referenced throughoutthis chapter. The published ICH guidelines are generally wellaccepted by regulators and industry alike, even outside thethree primary members (United States, EU, and Japan).There are cases where agencies will enforce requirementsbeyond those contained in the ICH guidelines; however, theguidelines are now often cited and accepted in a majority ofregulatory filings, and are utilized in development of newmedicinal products. The ultimate impact of ICH will bejudged in the coming years; however, it is already clear thatthis joint cooperation between agencies and industry is a stepforward. We look forward to increased levels of industry=regulator cooperation in this area in the years to come.

IX. HELPFUL REFERENCES

The Worldwide Web is an invaluable tool allowing today’sscientists to keep up to date on current regulatory guidancedocuments, along with as draft proposals, which govern thepreparation of investigational and marketing applications aswell as postapproval changes. An excellent FDA website(http:==www.fda.gov=cder=guidance=index.htm) contains acomprehensive compilation of the agency guidance docu-ments, arranged by disciplines. This user-friendly compila-tion also includes the latest ICH guidelines. A similar link(http:==www.emea.eu.int=index=indexh1.htm) exists to cur-rent EU guidance documents. While guidances and regula-tions in other countries are sometimes more difficult to find,use of web search tools definitely improve the chances forobtaining the latest information from regulatory agencies,worldwide.

A second set of valuable resources are the official compen-dia, including the United States Pharmacopoeia= National

200 Curran

Page 216: Active Pharmaceutical Ingredients

Formulary (USP=NF), European Pharmacopoeia (Ph Eur orEP), Japanese Pharmacopoeia (JP), and other local compen-dia, of which the British Pharmacopoeia (BP) is generally mostcomprehensive. These compendia provide insight into theexpectations for control of APIs expected by their publishingcountry, and also generally include general chapters guidingtesting, validation, interpretation of results, etc. Most of thecompendia are now available via the web, but generallyrequire a license fee per user.

Regulatory Affairs 201

Page 217: Active Pharmaceutical Ingredients
Page 218: Active Pharmaceutical Ingredients

6

ValidationJAMES AGALLOCO

Agalloco & Associates, Inc., Belle Ineade, New Jersey, U.S.A.

PHIL DESANTIS

Schering-Plough Corp., Kenilworth, New Jersey, U.S.A.

I. History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203II. Definition of Validation . . . . . . . . . . . . . . . . . . . 205III. Regulations . . . . . . . . . . . . . . . . . . . . . . . . . . 205IV. Application of Validation . . . . . . . . . . . . . . . . . . 206V. Life Cycle Model . . . . . . . . . . . . . . . . . . . . . . . 206VI. Validation of New Products . . . . . . . . . . . . . . . . 206VII. Validation of Existing Products . . . . . . . . . . . . . . . 208VIII. Implementation . . . . . . . . . . . . . . . . . . . . . . . . 208IX. Bulk Pharmaceutical Chemical Validation . . . . . . . . 211X. In-Process Controls . . . . . . . . . . . . . . . . . . . . . 220XI. Cleaning Validation . . . . . . . . . . . . . . . . . . . . . 222XII. Computerized Systems . . . . . . . . . . . . . . . . . . . 225XIII. Procedures and Personnel . . . . . . . . . . . . . . . . . 225XIV. Validation of Sterile Bulk Production . . . . . . . . . . . 225XV. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . 231

References . . . . . . . . . . . . . . . . . . . . . . . . . . 231

I. HISTORY

Validation was initially introduced in the 1970s to the phar-maceutical industry as a means for more firmly establishingthe sterility of drug products where normal analytical meth-ods are wholly inadequate for that purpose. In followingyears, its application was extended to numerous other aspectsof pharmaceutical operations: water systems, environmentalcontrol, tablet,and capsule formulations, analytical methods,and computerized systems. Individuals working with bulkpharmaceutical chemicals (BPCs) were particularly reluctantto embrace validation as a necessary practice in their opera-tions. Industry apologists explained this lack of enthusiasm

203

Page 219: Active Pharmaceutical Ingredients

in terms of differences in facilities, equipment, technology,hygienic requirements, cleaning methodologies, operationalpractice, and numerous other aspects of disparity thatseemingly justified the recalcitrance of this segment of theindustry. This view was widespread in the BPC industrythrough the end of the1980s.

The extension of the concepts that have made validationsuch an integral part of practices across the healthcare indus-try to the production of BPCs seems obvious in retrospect.Yet, for many years there existed a general reluctance tointroduce validation into BPC activities. While there weresome modest efforts, it was not until some time after the bio-technology industry became technically and commerciallyviable that any significant effort was initiated. The produc-tion of biotech products for registration in the United Statesrequires the approval of FDA’s Center for Biological Evalua-tion and Research (CBER). Center for Biological Evaluationand Research required extensive validation of fermentation,isolation and purification processes utilized in the preparationof biologicals (1). An objective comparison of BPC operationsrelative to those performed in the early stages of biologicalswould reveal minimal differences. The production methodolo-gies for many classical BPCs, e.g., penicillins, cephalosporins,and tetracyclines are nearly indistinguishable from thoseutilized to prepare tPA, EPO, and other biologicals. With thisrealization, the advent of validation for BPCs was apparent toall, and was increasingly imposed upon the industry.

In 1990, the U.S. Pharmaceutical Manufacturers Asso-ciation (now called PhRMA) formed a committee to defineBPC validation concepts (2). This committee’s efforts culmi-nated in 1995 when they issued their finished draft. Thisdocument served as a guide to the authors in the developmentof this chapter. Of necessity, considerable clarification andexpansion of the material contained has been necessary tocomplete this effort.

In the late 1990s, a new term started to appear, first inEurope, but soon it spread across the entire industry—activepharmaceutical ingredients or APIs. Those who first usedthe new term suggested that it was synonymous with bulk

204 Agalloco and Desantis

Page 220: Active Pharmaceutical Ingredients

pharmaceutical chemicals or BPCs. Since that time, it hasbecome increasingly common in the industry speak only ofAPIs. A part of the rationale for this initiative has been voicedas a move towards harmonization. The authors of this chapterdo not agree with this change in terminology, as there arenumerous bulk pharmaceutical chemicals that have no meta-bolic activity. Many pharmacologically inactive materials areproduced within the industry using facilities, equipment, andmethodologies identical to that employed for the so-calledAPIs, yet with the advent of this new catch phrase are to beseemingly ignored. Our use of the term bulk pharmaceuticalchemical is deliberate and is intended to embrace both activemoieties, and therapeutically inert materials used as excipi-ents, processing aids, and other materials.

The official requirement for validation of BPC processeswas formally established in Guidance for Industry, Q7A GoodManufacturing Practice Guidance for Active PharmaceuticalIngredients (3). This was the result of a multiyear effort bythe International Conference on Harmonization (ICH), whichresulted in this harmonized guidance document. This gui-dance document addresses the subject of validation briefly,and employs the same definition FDA has adopted for otherprocesses (see next paragraph). This chapter provides recom-mendations for validation consistent with the Q7A guidance.

II. DEFINITION OF VALIDATION

There are innumerable definitions of validation that havebeen written over the nearly 30 years since its appearance inthe pharmaceutical industry. Rather than foster newdefinitions with the context of this chapter, the authors havechosen to draw upon some of the more widely quoted defini-tions. The FDA defines process validation as: ‘‘Process valida-tion is establishing documented evidences, which provides ahigh degree of assurance that a specific process will consis-tently produce a product meeting its predetermined specifica-tions and quality characteristics’’ (4). This definition isreferred to in FDA’s subsequent guidance specific for BPCs (5).

Validation 205

Page 221: Active Pharmaceutical Ingredients

III. REGULATIONS

Regulations specific to the control BPCs are a relatively newconcept; for many years FDA’s policy was to apply a limitedenforcement of the subpart 211 regulations for finished phar-maceuticals (6). In recent tears, FDA has endeavored to har-monize its approach to BPC regulation with the rest of theworld, and has issued a guidance document that draws heavilyon subpart 211 (7). This effort followed the issuance of a phar-maceutical inspection convention document that addressedthe same subject in a different format (8).

IV. APPLICATION OF VALIDATION

Some discussion of validation approaches utilized for BPCs isessential to following this chapter. The approaches for BPCsare essentially the same as those utilized for other processesand systems. This discussion serves to highlight the nuancesof validation as they apply to BPCs.

V. LIFE CYCLE MODEL

Contemporary approaches to the validation of virtually anytype of process or system utilize the ‘‘life cycle’’ concept (9).The ‘‘life cycle’’ concept entails consideration of process or sys-tem design, development, operation, and maintenance at theonset. Use of the life cycle helps to provide a system that meetsregulatory requirements, but is also rapidly placed into service,operates reliably, andeasilymaintained.While the ‘‘life cycle’’ isbest suited to new products, processes, or systems, it certainlyhas applicability for existing systems as well. Existing systemsthat have never been previously validated can be reviewedagainst the same validation criteria that would be imposed fornew systems.While these systems are likely to be deficientwithregard to current requirements, the ‘‘life cycle’’model provides ameans for upgrading their programs to be on a par with newlydeveloped systems. This is especially important for bulk phar-maceutical chemicals given that the validation of these pro-cesses has lagged behind many of the other areas of the

206 Agalloco and Desantis

Page 222: Active Pharmaceutical Ingredients

industry,where validationhasalreadybeen instituted. It is per-haps safe to say that the first validation efforts to be utilized forBPCs will likely be retrospective ones, following the existingsystem path to enter the ‘‘life cycle’’ model.

VI. VALIDATION OF NEW PRODUCTS

The validation of a new BPC entails practices that parallelthose utilized for the introduction of a new pharmaceuticalformulation. Thus, a large part of the initial validation effortmust be linked to the developmental activities that precedecommercial-scale operation. The similarity is such thataspects of reaction, and purification methodologies shouldbe as similar as possible given of course the difference inthe scale of the equipment utilized in the commercial facil-ities. Any differences between the BPC process utilized forthe formulation batches used to establish clinical efficacyand the commercial material must be closely evaluated andtheir impact on the BPC products: chemistry, purity profile,stability, crystal morphology, and other key attributes.

The developmental laboratory has the responsibility fordetermining optimal reaction conditions including time,temperatures, raw material purity, molar ratios, solventselection, crystallization method, wash volume, drying con-ditions, etc. Of primary concern is the identification of criti-cal control parameters, that is to say those that impactquality, purity, safety, and efficacy. The concerns to beaddressed in any individual BPC validation program areof course unique to that process, the inclusion or exclusionof any single factor as a consideration in BPC validation isan arbitrary one determined by the authors. Chemical reac-tions are among the more complex processes to be subjectedto validation and the number of critical factors in even asingle reaction can be quite extensive. The amount of infor-mation which must be generated during development tosupport a validated BPC process is correspondingly exten-sive. The necessary information can be assembled into atechnology transfer document that conveys the collectedexperience gained during development to those responsible

Validation 207

Page 223: Active Pharmaceutical Ingredients

for the commercial production of the BPC. The success of adevelopmental organization is better assessed by the qualityof the information they convey to document their effortsthan it is by the sophistication of the chemistry utilizedto make the BPC. The technology transfer document islikely to be of central interest to FDA inspectors duringthe conduct of a preapproval inspection of the facility priorto approval (10).

VII. VALIDATION OF EXISTING PRODUCTS

At the time this is written, validation of bulk pharmaceuticalchemicals is a still relatively new concept for the industry toaddress. As such the vast majority of BPC products have beenon the market without any significant validation in place. Asa consequence, the first efforts to validate these products willundoubtedly employ retrospective methods. The trending ofresults derived from in-process and release testing of theseproducts and processes will serve as the basis for theseefforts. Given the FDA’s general dissatisfaction with retro-spective approaches, it is doubtful that these early efforts willremain the only approach utilized. The use of either pros-pective (in which three batches must be produced before theprocess can be considered validated and any of materialreleased for sale) or concurrent (in which individual batchesare released while continuing to accumulate data towards athree batch validation) approaches are certainly acceptable,a decision to use those approaches while raising less regula-tory concerns will also require a longer time to execute anda larger resource commitment.

The establishment of priorities for validation of a largenumber of BPC processes generally follows economic con-cerns, with those products that provide the largest contribu-tion to the firm’s profitability being the initial focus ofactivity. Regardless of how the first validation efforts werecompleted, the adoption of the ‘‘life cycle’’ model for maintain-ing products in a validated state is becoming increasinglywidespread.

208 Agalloco and Desantis

Page 224: Active Pharmaceutical Ingredients

VIII. IMPLEMENTATION

The validation of any process or product relies upon severalsupportive activities. Validation in the absence of these activ-ities has only minimal utility, as it is only through the inte-gration of these other practices that meaningful validationcan be accomplished. Several of these activities are defined inCGMP regulations while others are an integral part of a com-pany’s organization structure or are closely associated with‘‘validation’’ itself (11).

Equipment calibration—The process of confirming theaccuracy and precision of all measurements, instruments,etc. to ensure that the measured variable is being accuratelymonitored. Calibration includes demonstration of confor-mance to applicable national standards such as NIST, DIN,or BS for all key parameters. This is a universal CGMPrequirement across the globe.

Equipment qualification—An outgrowth of ‘‘validation’’that focuses on equipment related aspects. There is norequirement for a formal separation of the activity into dis-tinct elements, such as installation and operational qualifica-tion. It has become increasingly common in recent years tocombine these activities under a single effort. For the sakeof those who still separate the activities individual descrip-tions have been provided:

Installation qualification—Documentation that theequipment was manufactured and installed in accordancewith the intended design. This is essentially an audit of theinstallation against the equipment specifications and facilitydrawings.

Operational qualification—Confirmation that the equip-ment performs as intended entails evaluation of performancecapabilities. It incorporates measurements of speeds, pres-sures, and other parameters.

Process development—The development of products andprocesses, as well as the modification of existing processes,should be conducted to provide documented evidence of thesuitability of all critical process parameters and operatingranges. This effort serves as a baseline for all product validation

Validation 209

Page 225: Active Pharmaceutical Ingredients

activities. The integration of development into commercial-scale operations became a requirement with the advent of theFDA’s preapproval inspection program (10). The importanceof well-documented developmental activities to support subse-quent commercial-scale production is essential in the valida-tion of BPCs. It is customary for many unit operations(reactions, separations, catalyst reuse, solvent reuse, etc.) tobe initially confirmed on a laboratory or pilot scale, prior totheir eventual ‘‘validation’’ on a commercial process scale.

Process documentation—An often overlooked activitywherein the results of the development effort are delineatedin sufficient detail in process documentation so that the varia-tions in the process as a result of inadequately defined proce-dures are eliminated. While master batch records have longbeen a CGMP requirement, their adequacy is essential tothe maintenance of a validated state.

Performance qualification (testing)—That portion of theoverall ‘‘validation’’ program that deals specifically with theevaluation (validation) of the process. It includes the protocoldevelopment, data acquisition, report preparation, and therequisite approvals. In the distant past this activity was con-sidered ‘‘process validation,’’ but over the years the industryhas come to realize that ‘‘validation’’ encompasses a broaderspectrum of activities and continued use of the word ‘‘process’’is limiting.

Change control—A CGMP requirement that mandatesthe formal evaluation of the consequences of change to pro-ducts, processes or equipment. At least two distinct types ofchange control exist because of the different disciplines thatare central to the evaluation of each (12):

Process change control—A system whereby changes tothe process are carefully planned, implemented, evaluated,and documented to assure that product quality can be main-tained during the change process. This type of change controlis the province of the developmental scientist and productionpersonnel.

Equipment change control—A mechanism to monitorchange to previously qualified and=or validated equipmentto ensure that planned or unplanned repairs and modifica-

210 Agalloco and Desantis

Page 226: Active Pharmaceutical Ingredients

tions have no adverse impact on the equipment’s abilityto execute its intended task. This procedure usually entailsclose coordination with the maintenance and engineeringdepartments.

IX. BULK PHARMACEUTICAL CHEMICALVALIDATION

The focus of this chapter is bulk pharmaceutical chemicalvalidation. Aside from the history section, the informationpresented to this point would apply to most any type ofprocess. That commonality with other older validation effortsis deliberate. Bulk pharmaceutical chemical validation isunique, only to the extent that BPCs are unique. The under-lying maxims of success for validation (the knowledge andunderstanding of the scientific basis upon which the equip-ment or process is based) are universal. Mastery of the overallapproach equips one to effectively employ those concepts in avariety of settings. Some knowledge of the key concerns in theproduction of BPCs is essential to understanding how thevalidation of their preparation should be carried out.

Unit operations—BPCs are the result of a series ofchemical reactions in which materials, called reactants, arebrought together under appropriate conditions whereby thereaction takes place and the reaction product is formed.Under even the most ideal circumstances, the desired productmust be separated from unreacted raw materials, byproducts,solvents, and processing aids before it can be utilized infurther processing. In the analysis of these processes, chemi-cal engineers have found it convenient to divide the overallprocess into a series of unit operations (some of which arephysical in nature, while others ‘‘reactions’’ are chemical innature). The unit operations approach is beneficial becausea complex many-step process can be separated and betterunderstood as a series of simpler activities (unit operations)that are more easily interpreted.

Among the more common unit operations are mixing,heating, drying, absorption, distillation, condensation, extrac-

Validation 211

Page 227: Active Pharmaceutical Ingredients

tion, precipitation, crystallization, filtration, and dissolution.There are other less common unit operations, but the moreimportant aspect is the subdivision of a lengthy process intosmaller and more readily understood segments. The benefitsto be gained from this approach are obvious, once the under-lying principles are understood for a specific unit operation,those concepts can be reapplied in other steps or processeswhere that same unit operation is employed. In the validationof BPC processes, the ability to use standardized methods foreach unit operation can make what would otherwise be animpossible task into a manageable one. The unit operationapproach is of such utility that it has been applied in pharma-ceutical dosage form manufacturing as well, where the samebasic procedures are often encountered, i.e., mixing, milling,filtration, etc.

Physical parameters—A concern that has been some-times neglected in the preparation of BPCs relates to the con-trol of physical parameters of the end product material. Oftenthe focus of BPC development and processing is on chemicalpurity and yield, as those aspects tend to have the greatesteconomic significance. There is relative indifference to physi-cal parameters such as size, shape, and density compared tothe seemingly more important concerns such as potency,impurity levels, and process yield. The authors have observednumerous situations where this inattention has resulted inprocessing problems at the dosage form manufacturing stage.In each instance, it was often the case that the physical para-meters of the end product had been virtually ignored in defer-ence to concerns over chemical purity. The FDA’s preapprovalinspection initiative indicated an awareness that these con-cerns have come to their attention during the course ofNDA reviews and inspections (10).

The most extreme circumstances where physical para-meters are of critical importance are for those materialswhere different crystalline forms are possible. The differentpolymorphs may have decidedly different characteristics withregard to crystal shape, size, and most importantly solutioncharacteristics. Many important pharmaceutical chemicalsexist in more than one crystalline form, and the manufacturer

212 Agalloco and Desantis

Page 228: Active Pharmaceutical Ingredients

must insure that only the desired form is being produced. Oneof the major concerns voiced by regulators is the potentialhazard in using brokered active ingredients (5). The abilityto match the purity profile of a BPC is not sufficient if thecrystallization is from a different solvent system or at differ-ent conditions. An entirely different material may result, withprofoundly different pharmacological properties. The absenceof detailed information on the isolation process used maycause difficulties should the real source of the material (thebroker’s supplier) change.

Chemical purity—Central to the preparation of BPCs areissues relative to the purity of the desired material. Untilrecently, the only concern was whether the material metthe minimum potency requirements. A typical requirementwould be a minimum potency specification of 98%. Any lotthat had an assay higher than 98% would be acceptable.Awareness that the small amount of material that is not thedesired molecule could cause adverse reactions led to theestablishment of purity profiles for the molecule. Using a pur-ity profile approach mandates that the firm identify the impu-rities present. Current FDA expectations are that firmsshould characterize all impurities that comprise more than0.1% of the material and perform toxicity testing on anyimpurity that is at a concentration higher than 0.5% (5).The establishment of a purity profile for a molecular entityassures that process changes, which might result in a changein the byproducts and other materials isolated with thedesired material, do not impact the safety and efficacy ofthe final product.

Analytical methods—As with other types of productvalidation activities, BPC validation cannot proceed withoutvalidated analytical methods. The most significant differencein the validation of BPCs is the number of analytical methodsthat must be addressed. Analytical methods are needed foreach stage intermediate, identifying and quantifying themajor byproducts at each stage as well as the desired chemi-cal moiety. Clearly, the scope of the analytical method valida-tion for BPCs represents a larger effort than is normallyassociated with process validation activities. A comprehensive

Validation 213

Page 229: Active Pharmaceutical Ingredients

review of analytical method validation can be found elsewherein this volume.

Facilities—BPC facilities are vastly different from mostother types of facilities in the pharmaceutical industry. Theequipment is designed for specialized procedures and as suchbears little resemblance to those that might be found in adosage form facility. Most BPC equipment requires a broaderrange of utilities and a seeming maze of piping to performproperly. Chemical reactions are sometimes performed attemperatures in excess of 120�C or less than 0�C andrequired specializing heat transfer fluids to maintain thosetemperatures.

Many reactions utilize solvents as reaction substrates orin the isolation of the materials. These solvents are sometimesintroduced via piping systems that supply the various piecesof equipment. Distribution systems for compressed gases usedeither in the reaction or to inert the equipment are also com-mon. In many older BPC facilities, it is common to see multi-ple vessels at different elevations arranged around an openbay. In these facilities, several different chemical reactionsmight be underway in different vessels for different pro-ducts at the same time. In a dosage form facility, this typeof arrangement would be viewed with some skepticism. InBPC production, the reactions and unit operations take placewithin completely closed equipment, minimizing the potentialfor cross-contamination. The difference between BPC anddosage form facilities is most evident in warmer climates. Inthese areas, the BPC facility may be little more than struc-tural support for the equipment and staging areas for mate-rial, with no surrounding building. In effect, the equipmentis outside, fully exposed to the environment. For certainBPC processes such as solvent recovery and hydrogenationvessels, the equipment is located outside in even northern cli-mates either because of sheer size or safety concerns. Thesetypes of arrangements are not typical for the last step inthe synthesis. Isolation of the completed BPC is usually per-formed in rooms specifically designed for that purpose.

Pure rooms—In the preparation of BPCs, it is common forthe last step in the process to be completed in an environment

214 Agalloco and Desantis

Page 230: Active Pharmaceutical Ingredients

far different from that in which the rest of the synthesis isperformed. The term ‘‘pure room’’ is used loosely, there areno regulatory requirements for these rooms and the actualterminology varies considerably from firm to firm. Even with-out regulatory impetus, some firms have gone so far as toclassify their pure rooms at Class 100,000 (EU Class D) orbetter (13,14). After the crystallization of the BPC, it isimportant to protect the product from airborne particulates,and other foreign matter that might end up in the finishedmaterial. For this reason it is common in many companiesto perform a filtration of the active material while still insolution. The filtration removes particulates that may haveaccumulated in the material up to that point. After the filter,the solution is introduced into the crystallizer in the pureroom. The room itself is designed to minimize the opportunityfor introduction of contaminants into the bulk material andmay or may not be a classified environment. The crystallizeris often subjected to extraordinary cleaning before the start ofthe process to ensure its suitability for the final bulk isola-tion. Following the crystallization, the BPC is centrifuged,washed, dried, milled, and packaged in the pure room. Itshould be noted that BPC processes which use pure roomsare not intended to be sterile, the production of sterile BPCsrequires a much higher level of control over the environment,equipment, and methodologies and is described more fullylater in this chapter.

Qualification of equipment—The qualification of BPCprocess equipment including reaction vessels, receivers, crys-tallizers, centrifuges, dryers, filters, distillation columns, sol-vent distribution systems, etc. is a well-defined activity. Whilethis equipment is somewhat different in design and operatingfeatures, than the dosage form equipment that has been thesubject of the majority of papers on the subject, the same gen-eral principles apply. Reaction vessels, receivers, and crystal-lizers differ only minimally from formulation and water forinjection tanks. Some BPC dryers are identical to those uti-lized in tablet departments. Solvent distribution systemsare piping systems and may resemble WFI distribution sys-tems. Some pieces of equipment such as distillation columns

Validation 215

Page 231: Active Pharmaceutical Ingredients

and continuous reactors may not have counterparts in thedosage form side, but an understanding of the objectives ofthe equipment qualification should make the development ofsuitable protocols straightforward.

Configuration confirmation—In multipurpose BPC facil-ities, the fixed equipment installed may be configured differ-ently for different reactions. In these facilities, campaigns ofone reaction may be followed by a reaction for a different pro-duct after a change in configuration. Putting aside cleaningconsiderations for a later portion of the chapter, verificationof the systems configuration should be performed. In effect,the reaction train must be requalified at the start of each cam-paign to insure that the proper arrangement of valves, trans-fer lines, instruments, and other items are established for theprocess to be introduced. Some firms run a water or solventbatch, which simulates the process to verify that the properconnections are in place and that there are no leaks in the sys-tem. Following the trial batch, the system is then readied foruse with the solvents that will be utilized in the process.

Environmental control—The usual concerns relative tothe environment in which the production activities are per-formed are not as significant in BPC manufacturing as theyare for the preparation of pharmaceutical dosage forms. Theintroduction of microbial or particulate contaminants at earlystages of the process is unlikely to be of significance. BPCreactions utilize high temperatures, extremes of pH, andaggressive solvents that can minimize the impact of anymicrobial contamination. Filtration is a frequent part ofBPC processing in the form of carbon treatments and otherunit operations whose intent is to remove unwanted bypro-ducts, reactants, and solvents. In the course of these mea-sures, incidental particulate contamination is also removed.The use of ‘‘pure rooms’’ as outlined earlier serves to minimizecontamination at the last step.

Worker safety—The safety of the personnel who work inthe facility is always a major concern. Exposure to toxic sub-stances is greatest when the operator is adding materials to orremoving materials from the equipment. The use of air extrac-tion equipment, isolation technology, automated handling, and

216 Agalloco and Desantis

Page 232: Active Pharmaceutical Ingredients

other means for minimizing human contact with toxic materi-als is nearly universal. The assessment of worker safetyshould also embrace exposure to vapor phase hazards, andleak testing of process trains should be performed wherehazardous gases are present. Validation of the effectivenessof this equipment is not mandatory from a CGMP perspective,but is certainly beneficial.

Process water—The water used in BPC production isusually deionized water through the early process stages. Ifthe product is isolated from a water solution in its last step,then a compendial grade of water, purified water or WFImay be utilized depending upon subsequent steps in dosagemanufacture and the final use of the product. Cleaning ofequipment can be performed with city water, provided thelast rinse of the equipment is with the same water utilizedin the process step. The validation of water systems has beenwell documented in the literature (15,16).

Process gases—Some BPC reactions utilize gases as reac-tants, or are performed under a gas blanket. The system maystart at either a large high-pressure bulk storage tank or froma bank of gas cylinders. Attention should be paid during theinstallation of the system to assure that the materials of con-struction utilized in the system are compatible with the gasbeing handled. Distribution systems for these gases requirequalification, but their similarity to gas distribution systemsused in dosage form facilities means that the basic approachis well defined in the literature. For safety considerations par-ticular attention should to be paid to proper identification ofprocess gas lines throughout the facility (see following para-graph).

Compressed air—Air which is classified as breathableshould receive an intensive qualification effort especially withregard to the verification of ‘‘as-built’’ drawings, confirmationof proper identification, as well as any safety- and purity-related issues. The emphasis given to breathable air is dueto the number of unnecessary deaths, which have occurredin the industry as a consequence of misidentified gas lines.Where air is utilized as a reactant in a BPC operation, itshould be treated as described previously under process

Validation 217

Page 233: Active Pharmaceutical Ingredients

gases. Instrument air requires the least intensive effort, asthe adequacy of the installation can be often confirmedindirectly during the calibration and qualification of the pro-cess instrumentation. A single compressed air system couldserve as the source for more than one of these air systemssimultaneously. In this instance, the advice provided for themost critical application is appropriate throughout.

Jacket services—It is common in BPC facilities, espe-cially those which are reconfigured frequently to accommo-date the production of different materials, to have eachmajor vessel equipped with identical utilities, such as chilledwater, plant steam, compressed air, and coolant. The use ofidentical utility configurations on the vessels maximizes theflexibility of the facility, reduces the potential for operatorerror, and simplifies the design of the facility. The control sys-tems for these jacket services on the vessels would also beidentical. Under these circumstances, the qualification effortis greatly simplified through the use of identical require-ments.

Solvent distribution—Many facilities use one or moresolvents repetitively. In these instances, the installation of adedicated distribution system for the solvent to the varioususe points can be justifiable. These systems may be lengthylines from the bulk storage area (tank farm) to the variouslocations in the facility where the solvent is required. In somecases, a chilled solvent system may be present to providechilled washes for use in centrifugation. Depending uponthe solvent, specialized piping or gasket materials may benecessary to avoid leaks or corrosion of the system. Qualifica-tion of these distribution systems is easily accomplished.

Solvent recovery and reuse—The reuse of organicsolvents in a BPC system is widespread, especially giventhe increased cost of these materials and the environmentaldifficulties sometimes associated with their proper disposal.This reuse is achieved through defined procedures for therecovery of the solvents from distillates, extractions, andspent mother liquors. Where recovered solvents are utilizedin the production of a BPC, the validation of the recovery pro-cess is strongly recommended. The validation of the recovery

218 Agalloco and Desantis

Page 234: Active Pharmaceutical Ingredients

process would include all steps in the process, and confirm theacceptability of the recovered solvent in the processes it willbe utilized in. The validation of the use of recovered solventscould be a part of the development of the process. Repeatedrecycling of solvents could result in the concentration of traceimpurities that could adversely affect reaction chemistry. Atthe very least, recovered solvents should be subjected torelease testing and shown to be comparable to fresh solvent.The complexities associated with the validation and reuse ofrecovered solvents should not be overlooked.

Multiple crops—In the crystallization of some BPCs,multiple crops are sometimes utilized to maximize theamount of material isolated. Even where the cost of the mate-rials being isolated is not high, the ability to increase theoverall yield through the preparation of second, third, or evenfourth crops is frequently a routine part of the process. Arelated technique is to recycle the mother liquors withoutadditional treatment from the crystallization to the beginningof the process. Whether through multiple crops or recycling ofthe mother liquor, both of these processes result in the con-centration and=or retention of impurities. The validation ofthese practices must be a part of the development effort forthe process, and reconfirmed on the commercial scale.

Catalyst reuse—Precious and semiprecious metals andother materials are often utilized as catalysts in the conductof certain chemical reactions: e.g., hydrogenation. While thequantity of catalyst required in any particular reaction isquite low, the cost of these metals is such that recovery ismandated. As the amount of catalyst required to supportthe reaction is generally supplied in excess it is frequentlypossible to return the catalyst to the start of the process stepwithout loss in effective yield. The reuse of the catalyst inthis manner must be supported by appropriate developmentwork.

Waste treatment—The nature of the materials, bypro-ducts, and solvents utilized in the preparation of BPCs ulti-mately results in any number of waste treatment problems.The validation of these treatments is certainly not a CGMPrequired activity. Nevertheless, consideration should be given

Validation 219

Page 235: Active Pharmaceutical Ingredients

to those activities to insure their reliability. Such efforts canaid in attaining environmental approval for the facility.

X. IN-PROCESS CONTROLS

Bulk pharmaceutical chemicals resemble other types ofproducts validated in the pharmaceutical industry in thatthey utilize various in-process controls to support and monitorthe process through its execution. Typical controls that mightbe a part of a BPC process include material specifications.

Material specifications—The controls of reactants,solvents, intermediates, and finished materials employ formalspecifications for key parameters. The importance of thesecontrols increases towards the end of the synthesis and anyof the controls that follow the BPC step are certainly impor-tant enough that the efficacy of limits set for these controlsshould be a major part of the developmental process. Fore-most among the considerations in the latter process stepsshould be the impurity profile of the key intermediates (seefollowing paragraph). Physical parameters (size, shape, crys-talline form, bulk density, static charge, etc.) of the finishedBPC are sometimes considered less important than chemicalpurity. When the BPC is formulated in a solid or semisoliddosage form, these physical parameters may assume fargreater significance.

Purity profiles—Within the specification parameters,prominence is often given to the establishment of purity pro-files for the key intermediates and finished goods. The FDAmandates the identification of all impurities with a concentra-tion greater than 0.1% and generation of safety and other cri-tical information for impurities at levels of 0.5% or higher (5).The establishment of purity profiles for the final BPCs pro-vides for confirmation of the safety of the active material. Itis often beneficial to establish purity profiles for intermedi-ates earlier in the synthesis to prevent the carryover ofimpurities to the finished BPC. The maintenance of the purityprofile mandates that a careful evaluation of process changesand potential alternate suppliers of solvents, raw materials,

220 Agalloco and Desantis

Page 236: Active Pharmaceutical Ingredients

intermediates, and BPCs be made. The analytical methoddevelopment and synthetic chemistry skills required to obtainthe necessary data on impurities meeting the FDA’s criteriaare substantial. These efforts are well rewarded in anexpanded knowledge of the process chemistry and analysisthat can assure the quality of the desired active moiety.

Vendor support to validation—A common practice inBPC production is the subcontracting of certain chemicalsteps to outside suppliers. As is the case with subcontractedproduction for dosage forms, the owner of the NDA or DMFmaintains responsibility for the validation of the processand must secure the cooperation of the subcontractor in theperformance of any supportive qualification=validation activ-ities. Agreement to this arrangement should be a preconditionto the awarding of the contract to the supplier.

Supplier quality evaluation and audits—Suppliers ofintermediates, reactants, solvents, and other materialsshould be subjected to the same types of evaluation utilizedfor other dosage forms. The extent of the assessment shouldvary with the importance of the material to the process. Pre-cedence would be given to those materials whose purity wouldhave an increased impact on the finished BPC. Where thematerial being produced by the vendor has direct impact onthe BPCs quality, as would be the case for chemical inter-mediates, a more intensive approach is required. Periodicaudits of these key suppliers should be a part of the overallquality assurance program.

Sampling plans—Obtaining samples of finished BPCs ortheir intermediates presents the same difficulties encoun-tered in the sampling of any similar material. When samplesare taken of powder or crystalline materials, questions regard-ing the uniformity of the material being sampled must beaddressed before the results of the sampling can be consideredmeaningful. Bulk pharmaceutical chemicals that are driedin rotary or fluidized bed dryers may be blended sufficientlyas a result of the drying process. However, where tray dryersare utilized, a final blending of the dried material may berequired before sampling for release to the next stage ofprocessing. In certain instances, an intermediate or finished

Validation 221

Page 237: Active Pharmaceutical Ingredients

material will not be isolated as a dry powder but will bereleased as a solution in an appropriate solvent. Under thesecircumstances, concerns regarding the sampling of the mate-rial are minimized.

Particle sizing—Milling and micronizing are commonactivities in the final stages of BPC manufacture. These pro-cedures are utilized where the BPC producer has committedto providing a particular particle size for use in the formula-tion. Given the importance of particle size in many finaldosage forms, where present these processes should be vali-dated. Control of the final particle size for finished BPCshould not rely on the milling=micronizing step alone. Controlover the crystallization procedure is generally necessary tominimize the variation in the material that is to be sized inthe mill. It should not be assumed, that the milling=microni-micronization procedure will be tolerant of a wide range ofmaterials and still provide a consistently sized finished BPCproduct. The uniformity of materials is sometimes improvedby passage through a particle sizing procedure or sifter, butthis step alone should not be considered sufficient to achievea uniform mix of the material prior to sampling (see priorparagraph).

Reprocessing—There is occasional need to reprocess anintermediate or finished BPC in order to alter its crystal size,reduce impurities, or otherwise recover off-specification mate-rial. Where these processes are utilized, their inclusion in thevalidation program is essential. FDA requirements on repro-cessing and reworking of materials require the validation ofany material reclaimed in this fashion. This is most readilyaccomplished as a part of the developmental process.

XI. CLEANING VALIDATION

A comprehensive discussion of cleaning validation is beyondthe scope of this chapter, the reader should refer to othersources on cleaning validation for details of this activity(17–19). Within the context of this chapter, only those aspectsof cleaning validation unique to BPC production will be

222 Agalloco and Desantis

Page 238: Active Pharmaceutical Ingredients

presented. Additional guidance can be found in FDA’s BPCInspection Guide (5).

Boil-outs—Commonly used to clean BPC equipment,boil-outs entail the introduction of the solvent (it could bewater) used in the just completed process, and heating it toreflux. The expectation is that the evaporation=condensationdensation will result in the dissolution of any residue on theequipment in the solvent. This will remove it from the inter-nal surfaces that are ordinarily inaccessible for direct clean-ing and thus clean them. Boil-outs are also utilized as one ofthe last steps in preparation of equipment for the start of aprocess or campaign.

Lot-to-lot cleaning—As the production of BPCs oftenrequires that solvents and materials with substantial toxicitymust be employed, cleaning of the equipment after completionof the process has the potential for exposure of the worker tothose materials. For this reason, it is common in BPC facil-ities to include some basic forms of waste treatment andequipment cleaning directly into the process in an effort tominimize worker exposure later on. In addition to these mea-sures, many processes include the reuse of equipment andretention of materials in the equipment without cleaning. Atypical instance would be leaving a heel in the centrifuge atthe completion of the batch, thereby eliminating cleaning ofthe centrifuge after each batch. The retention of the heel mustbe validated as it represents a portion of the first batch, whichmay now become a part of subsequent batches. In fact, eachbatch in the entire campaign is potentially mixed with mate-rial from every prior batch! In this manner, the amount ofcleaning required between batches of the same reaction stepwould be reduced. In those facilities, where a process trainis essentially dedicated to the same reaction step over a longperiod of time, the equipment and process are speciallydesigned to minimize batch-to-batch cleaning of the equip-ment. There are of course instances where the presence ofeven trace quantities of finished material at the start of thereaction may create an undesirable outcome, in those circum-stances the equipment must be cleaned after the completionof each batch. Sparkler and other filters used to recapture

Validation 223

Page 239: Active Pharmaceutical Ingredients

catalysts, activated carbon used for decolorization and bypro-ducts may require cleaning after every batch.

Campaigns—The production of a number of batchesof an identical synthesis in the same equipment is commonin the manufacture of BPCs. As mentioned earlier inrelation to the qualification of equipment, production in acampaign mode may require the partial reconfiguration ofthe equipment train to allow for a new campaign. Thismay be a reaction leading to the same or a different BPC.To allow for campaign usage, the extent of cleaning requiredwill generally be far greater than what is carried outbetween batches of the same process step. Cleaning limitsfor campaign cleaning are generally tighter than thoseapplied for batch-to-batch cleaning. It is beneficial in cam-paign cleaning to follow a defined plan for changeover fromone product to another.

Sampling for residuals—In order to determine whether apiece of equipment has been appropriately cleaned, samplingis performed. Here again, the particular nature of the BPCmaterials makes for a more difficult situation. In dosage formmanufacturing, relatively few of the materials likely to beretained on the surface of the equipment poses any substan-tial risk to the worker. In those dosage form processes wheretoxic or potent materials are handled, the design of the equip-ment with smooth surfaces, rounded corners, sanitary fit-tings, etc. reduces cleaning difficulty. The same equipmentdesign principles make sampling of pharmaceutical equip-ment relatively simple due to provisions for access and inspec-tion. The bulk of BPC equipment is designed to operate undermore aggressive conditions, and cannot always integrate thedesign features so commonly found in their pharmaceuticalcounterparts. Moreover, worker safety becomes a far greaterconcern, as the solvents and materials are not conducive todirect exposure to the employee. Sampling of BPC equipmentmay be restricted to fewer locations, and those locations aregenerally not in the most difficult to clean or ‘‘worst case’’locations. For this reason, the residual limits for BPCs mayneed to be far lower to accommodate the uncertainty of thesampling that can be performed.

224 Agalloco and Desantis

Page 240: Active Pharmaceutical Ingredients

XII. COMPUTERIZED SYSTEMS

The application of computerized systems in the pharmaceu-tical industry is perhaps greater in BPC processing than inany other. Distributed control systems (DCS) have been uti-lized for many years in the control and regulation of chemi-cal process plants. Their adaptation to BPC preparation isstraightforward. The validation of computerized systems inthe pharmaceutical industry has been extensively discussed,with the constant recognition that their extensive usage inBPC production was a given (20,21). Industry and regulatoryguidance having always recognized this fact, this chaptercould not hope to do justice to the subject which has filledseveral textbooks on its own. The reader is encouraged tofollow the recommended practices of PDA, PhRMA, andGAMP.

XIII. PROCEDURES AND PERSONNEL

Where computerized systems are not utilized for the execu-tion of the chemical synthesis, the chemical operator, follow-ing detailed batch records is responsible for the operation ofthe equipment. The batch records must provide for sufficientdetail to insure that the worker can safely and properly per-form the desired actions. In certain larger process trains,more than one operator will work simultaneously on the samebatch. Provided that their activities are closely integrated,there is little problem with this type of approach. The person-nel must be trained in their jobs and records of the trainingmust be retained by the firm.

XIV. VALIDATION OF STERILE BULKPRODUCTION

The preparation of BPCs, which must be sterile upon comple-tion of their synthesis and purification, is a common activityin the pharmaceutical industry and increasingly common inbiotech processes. The validation of sterile BPCs represents

Validation 225

Page 241: Active Pharmaceutical Ingredients

one of the more difficult activities in the entire spectrum ofvalidation. Not only must the final material meet all of thephysical and chemical requirements associated with otherBPCs, it must also be free of microorganisms, endotoxin,and particulates. In doing so, all of the considerations for vali-dation of BPCs outlined in this chapter must be addressed,with added concern for sterilization, environmental control,aseptic technique, and other subjects associated with the pro-duction of sterile products. The following sections addressthose issues relating to sterile BPCs that are somewhat differ-ent from either the validation of nonsterile BPC production orthe validation of other sterile materials.

A. Product Sterilization and Sterility Assurance

The predominant method of sterilization for BPCs is by mem-brane filtration. This filtrationwill require validation in accordwith regulatory expectations. Adaptations to the commonfilter validation methodologies may be required for certain sol-vents and=or antibiotic solutions. Subsequent to the filtrationstep, the succeeding unit operations must be carried out usingfacilities, equipment, and methods designed to prevent theingress of microorganisms. The remainder of this sectionreviews considerations relative to sterile BPC preparationunder these constraints.

B. Closed Systems

Central to understanding much of what is presented below isrecognition that BPCs, whether intended to be sterile or not,are primarily produced in closed systems inwhich the reaction,separation, and purification unit operations take place. A jointPDA=PhRMA task force has defined a ‘‘closed system’’ as:

‘‘system which is designed to prevent the ingress ofmicro-organisms. A ‘‘closed’’ system may be more accu-rately defined by characteristics of its operation than bya description of its physical attributes, as these will varyfrom system to system (22).’’

226 Agalloco and Desantis

Page 242: Active Pharmaceutical Ingredients

A ‘‘closed’’ system

� is sterilized in place or sterilized while closed prior touse;

� is pressure and=or vacuum tight to some predefinedleak rate;

� can be utilized for its intended purpose withoutbreach to the integrity of the system;

� can be adapted for fluid transfers in and=or out whilemaintaining asepsis;

� is connectable to other closed systems while maintain-ing integrity of all closed systems (e.g., rapid transferport, steamed connection, etc.);

� utilizes sterilizing filters, which are integrity testedand traceable to each product lot.

Closed systems provide for complete separation betweenthe environment in which personnel (uniformly accepted asthe primary source of contamination in aseptic environments)are located from that in the materials are processed. Theore-tically, if a sterile BPC could be processed in its entiretywithin closed systems, there would no possibility of microbialcontamination. In marked contrast to the ‘‘closed system’’ isthe ‘‘open system’’, perhaps best defined by what it is not.Essentially, an ‘‘open system’’ lacks one or more of thefeatures of a ‘‘closed’’ system, thus leaving it vulnerable tothe potential ingress of contamination. One substantial issueassociated with these definitions is establishing that a systemremains ‘‘closed’’ over the length of the production campaign.

Facilities—The production of sterile BPCs requires acomposite of design features drawn from both sterile dosageform and bulk pharmaceutical chemical production. Ceiling,walls, and floors are composed of materials that can be sub-jected to frequent cleaning with disinfectants. Pressure differ-entials are provided to prevent the ingress of contaminationfrom less clean areas into critical processing areas. In orderto perform the reactions and separations necessary to prepareand isolate the BPC, processing equipment not generallyassociated with aseptic environments must be introduced.Centrifuges and crystallizers must be adapted for use in an

Validation 227

Page 243: Active Pharmaceutical Ingredients

aseptic area. The finished facility is most certainly a hybrid,as compromises are inevitable to accommodate the essentialrequirements. The case can be made that if the productionsystems are perfectly ‘‘closed,’’ then concerns relative tofacility design required for asepsis would be lessened. Theauthors are aware of several sterile bulk production facilitiesin which only a small portion of the system is actually locatedin an aseptic environment. Certainly, ‘‘open systems’’ mustapproach the highest levels of aseptic design in order to besuccessful in operation.

Environmental classification—The environments inwhich sterile BPC production is executed can vary with thedegree of closure provided by the equipment. ‘‘Closed’’ sys-tems as described earlier have been successfully operated inClass 100,000 (EU Class D) or unclassified environments.Systems that are open are generally located within Class100 (EU Class A) where product is exposed, and surroundedby Class 10,000 (EU Class B or C).

Utilities—There is very little difference between utilitysystems for a sterile bulk plant and those found in a typicalBPC facility. The only differencesmight be utilities uncommonin aBPC plant such aswater for injection and clean steam. Thevalidation requirements for these systems have been welldefined in the literature and need little mention here.

Layout—The layout of a sterile bulk facility will again bea hybrid of those found in a conventional BPC plant and asterile dosage form facility. There will be nesting of classifiedenvironments, with critical activities performed in the areasof highest classification. Pressure differentials are employedbetween clean areas, and those adjacent, less clean areas.The design features are drawn primarily from the dosageform facility model with adaptations to accommodate thegenerally larger equipment required for bulk production.

Isolation technology—The use of isolators and closedsystems for the production of sterile bulks is strongly recom-mended. As with any aseptic process, the sterility assurancelevel associated with a sterile bulk material is closely relatedto the extent of direct human intervention with the material.Isolators and closed systems minimize the need for personnel

228 Agalloco and Desantis

Page 244: Active Pharmaceutical Ingredients

contact with critical surfaces and thus minimize the potentialfor contamination of the sterile materials from human-bornemicroorganisms. It should also be noted that isolationtechnology can be useful in the containment of potent com-pounds as many BPC intermediates and finished materialsoften are. Isolation technology is a rapidly evolving areaand the reader is encouraged to stay abreast of current devel-opments (23,24).

Sterilization in place—Closed systems such as processvessels, dryers, centrifuges, isolators, and other items shouldbe subjected to a validated sterilization procedure, whichassures that all internal surfaces have been rendered freeof microorganisms. Sterilization-in-place (SIP) proceduresreduce the number of aseptic manipulations necessary toready the equipment for use in the aseptic production pro-cesses and as such are considered preferable to aseptic assem-bly of systems from individually sterilized components (25).The SIP procedure should allow the system to maintainsterility until ready for use without aseptic manipulations.Sterilization-in-place procedures could employ steam, gas,dry heat, radiation, chemical agents, or other validateablesterilization procedure.

A brief overview of some of the various sterilization-in-place methods available and their validation follows:

Steam—Primarily utilized for systems composed of closedvessels, with interconnecting piping. It has some similarity toempty chamber studies in steam sterilizers. Important para-meters to confirm are appropriate time–temperature condi-tions throughout the system. Emphasis is placed on theremoval of air and condensate from the system, strict adher-ence to the defined sequence for the sterilization procedureand inclusion of methods for the protection of the systembetween sterilization and use.

Gas—Utilized for systems that cannot withstand eitherthe temperatures or pressures employed in steam sterilization.Critical parameters for sterilization are time, temperature,relative humidity, and gas concentration. Gases in wide-spread use include ethylene oxide, peracetic acid, and hydro-gen peroxide. Gas sterilization is most often encountered

Validation 229

Page 245: Active Pharmaceutical Ingredients

with isolator systems, freeze dryers, and other systems whichhave limited ability to hold pressure.

Dry heat—Employed in specialized systems where thepresence of high temperature for the process is commonplace,i.e., spray dryers, flash dryers, and similar equipment. Confir-mation of time–temperature conditions in the equipment iscritical to the validation.

Radiation—Radiation sterilization is most commonly uti-lized for flexible packaging components that can be sterilizedwhile closed prior to filling. The validation of radiation steri-lization relies on confirmation of the delivered dose to all por-tions of the materials, and confirmation of stability after thetreatment.

Chemical—Many of the strong acids, strong bases, che-mical solvents and other chemicals utilized in the preparationof sterile BPCs have the ability to reliably destroy micro-organisms. These materials because of the extreme pH, orother aspects of their chemical structure can effectively steri-lize processing equipment. As their use in the system will gen-erally mandate that the equipment surfaces can be exposed tothese materials for extended periods of time, their use as asterilizing method for the equipment is facilitated. Concentra-tion and duration of contact are the critical parameters thatmust be confirmed in the validation of these treatments.

Aseptic processing—The validation of the sterile bulkprocess follows the general approach described earlier fornonsterile bulks. The overall process can be divided into aseries of unit operations that can be addressed individuallyor in groups. This approach can be used equally well foraspects of the chemical reaction, purification, physical proces-sing (i.e., milling, sieving, etc.) or aspects related to sterilityassurance. A comprehensive treatment of validation methodsfor validation of aseptic processing for sterile bulks has beendeveloped by a joint PDA=PhRMA task force (22). This docu-ment embraces such aspects of the validation as: the use ofclosed or open systems for processing; materials to use inthe conduct of the simulation; sampling and testing of materi-als; duration of simulation, simulation size, campaign produc-tion, and acceptance criteria to be employed. Producers of

230 Agalloco and Desantis

Page 246: Active Pharmaceutical Ingredients

sterile bulks are already familiar with the contents of thisdocument, and the interested reader is encouraged to readthis guidance in its original context.

XV. CONCLUSION

This chapter has provided an outline of validation considera-tions relative to the production of bulk pharmaceutical chemi-cals. This is a subject that has only recently become of interestto the pharmaceutical community. The authors while familiarwith both validation and bulk pharmaceutical processinghave undoubtedly mentioned any number of issues whichmay not yet be embodied in validation protocols within oper-ating companies. We have included these issues to insure com-pleteness in the presentation, not to suggest that they beincluded in every validation effort. As time passes, the industrywill gain experience with the validation of BPCs and will per-haps exclude some of these issues, while including other aspectswe have not identified. Our intent in this effort has always beento integrate common validation practices with the uniqueaspects of bulk pharmaceutical manufacturing. By no meansdo we expect this to be the definitive effort on this complex sub-ject. The reader is encouraged to monitor industry and regula-tory developments relative to BPC validation, as substantialchanges in CGMP requirements for BPCs appear likely.

REFERENCES

1. Food and Drug Administration, 21 CFR, Part 610.

2. PhRMA Quality Control Bulk Pharmaceuticals WorkingGroup. PhRMA Guidelines for the Production, Packing,Repacking or Holding of Drug Substances. PharmaceuticalTechnology, Part 1, December 1995, Part 2, January 1996.

3. Food and Drug Administration. Guidance for Industry, Q7AGood Manufacturing Practice Guidance for Active Pharmaceu-tical Ingredients. 2001.

Validation 231

Page 247: Active Pharmaceutical Ingredients

4. Food and Drug Administration. Guideline on General Princi-ples on Validation. 1987.

5. Food and Drug Administration. Guide to Inspection of BulkPharmaceutical Chemicals. 1994.

6. Food and Drug Administration, 21 CFR, Part 211.

7. Food and Drug Administration. Guidance to Industry: Manu-facturing, Processing or Holding Active Pharmaceutical Ingre-dients. March 1998.

8. Pharmaceutical Inspection Convention. Internationally Har-monized Guide for Active Pharmaceutical Ingredients—GoodManufacturing Practice. September 1997.

9. Agallco J. The validation life cycle. J Parenteral Sci Technol1993; 47(3).

10. Food and Drug Administration. Guide to Inspections of OralSolid Dosage Forms Pre=Post Approval Issues for Develop-ment and Validation. January 1994.

11. Agalloco J Validation—yesterday, today and tomorrow.Proceedings of Parenteral Drug Association International Sym-posium. Basel, Switzerland: Parental Drug Association, 1993.

12. Agalloco J. Computer systems validation—staying current:change control. Pharm Technol 1990; 14(1).

12a. Agalloco J. personal communications, 1972–1990.

13. Federal Standard 209E. Airborne Cleanliness Classes inCleanrooms and Clean Zones, September 1992.

14. EU Guide to Good Manufacturing Practice for Medicinal Pro-ducts. Annex 1—Manufacture of Sterile Medicinal Products.

15. Meltzer T. Pharmaceutical Water Systems. Tall Oaks Books1996.

16. Artiss D. AWater Systems Validation. In: Carleton F, AgallocoJ, eds. Validation of Aseptic Pharmaceutical Processes. NewYork: Marcel-Dekker, 1986.

17. Agalloco J. Points to consider in the validation of equipmentcleaning procedures. J Parenteral Sci Technol 1992; 46(5).

232 Agalloco and Desantis

Page 248: Active Pharmaceutical Ingredients

18. Madsen R, Agalloco J, et al. Points to consider for cleaningvalidation. PDA J Pharmaceutical Sci Technol 1998;52(6)(suppl). PDA Technical Report #29.

19. Voss J, et al. Cleaning and cleaning validation: a biotechnologyperspective. PDA, 1995.

20. Harris J, et al. Validation concepts for computer systems usedin the manufacture of drug products. Proceedings: Conceptsand Principles for the Validation of Computer Systems in theManufacture and Control of Drug Products, PharmaceuticalManufacturers Association, 1986.

21. Kemper C, et al. Validation of computer-related systems. PDAJ Pharmaceutical Sci Technol 1995; 49(1)(suppl). PDA Techni-cal Report #18.

22. Agalloco J, Lazar M, et al. Process simulation testing for sterilebulk pharmaceutical chemicals. PDA J Pharmaceutical SciTechnol 1998; 52(1). PDA Technical Report #28.

23. Akers J, Wagner C. Isolation Technology: Application in thePharmaceutical and Biotechnology Industries. Interpharm1995.

24. Coles T. Isolation Technology—a Practical Guide. Interpharm1998.

25. Agalloco J. Sterilization in place technology and validation. In:Agalloco J, Carelton FJ, eds. Validation of Pharmaceutical Pro-cesses: Sterile Products. Chapter. New York: Marcel-Dekker, 1998.

Validation 233

Page 249: Active Pharmaceutical Ingredients
Page 250: Active Pharmaceutical Ingredients

7

Quality Assurance and ControlMICHAEL C. VANDERZWAN

Pharmaceutical Technical, Roche Pharmaceuticals, Basel, Switzerland

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 235II. Defining and Assuring the Quality of the Active

Pharmaceutical Ingredient . . . . . . . . . . . . . . . . . . 240III. The Regulations for Quality . . . . . . . . . . . . . . . . . 245IV. The Quality Control and Quality Assurance Department 273

Appendix A . . . . . . . . . . . . . . . . . . . . . . . . . . . 280

I. INTRODUCTION

The quality of active pharmaceutical ingredients (APIs) isdefined as meeting the appropriate specifications for theAPI and being produced in a facility compliant with ICHguidelines ‘‘Q7A’’ and FDA’s current good manufacturingpractices (cGMPs) regulations. Most countries regulate themanufacture of APIs. These regulations require a total sys-tems approach to assuring an API has the appropriate levelof quality. All components in this system must be properlydesigned, validated, maintained, and operated to allow themanufacturer to assure the API consistently meets qualityrequirements. The general components of the system arethe process, facilities, and the people. This chapter concernsthese components, as well as the product quality itself, theregulations, and the quality management (QM) department.

235

Page 251: Active Pharmaceutical Ingredients

A. The Product

The quality of an API is determined by two factors: its confor-mance to pre-established specifications and whether it is pro-duced according to a documented validated process in a cGMPcompliant facility.

The API must possess appropriate chemical andphysical attributes to assure that it delivers the intendedpharmacological effect. The chemical attributes describe theappropriate purity and impurity limits. Impurity specifica-tions are established from clinical toxicological studies andare also based on reasonable minimums expected from regu-latory authorities and consumers. The physical attributesdescribe the necessary characteristics for reliable pharmaceu-tical processing into final dosage forms. These attributes aredetermined by empirical evidence from formulation trials toproduce uniform and stable dosage forms of adequate bioa-vailability.

B. The Process

The quality of the API is designed into the molecule throughthe development of the full manufacturing process, from thelaboratory scale synthetic process through to end product.The synthetic process must be designed to minimize impuri-ties, especially those that prove difficult to remove in the laststep. Thus, through effective process development, yields aremaximized, waste is minimized, and impurities are notformed, eliminated, or certainly minimized. The specific con-trols used by the developmental chemist to produce thehigh-yield, high-quality product must be documented; thisdocumentation forms the basis for the proof of concept andfor the validation report. In nearly all countries today, regula-tory authorities require the API to be produced from a docu-mented process that reliably meets all appropriatespecifications. This was strengthened by the issuance andadoption of the International Conference on HarmonizationTripartite Guideline of Q7A ‘‘Good Manufacturing PracticeGuide for APIs.’’ The European Union, the Japanese Ministry

236 VanderZwan

Page 252: Active Pharmaceutical Ingredients

of Health and the United States Food & Drug Administrationadopted the guide.

C. The Facilities

The facilities in which APIs are produced are also addressedin this chapter because a component of quality of an API isthat it be produced in cGMP-compliant facilities. Those com-ponents of the facility governed by cGMP are therefore partof this chapter. The essence of cGMP for facilities or, for thatmatter, any aspect of API manufacture is that the facility per-forms as designed to assure the quality of the product.Further, the performance characteristic must be documented,and management must demonstrate the facility continuallyperforms as designed. Performance control monitoring, pre-ventative maintenance, and carefully controlled and approvedrepairs or changes to facility components are all consideredpart of assuring quality of APIs.

D. The People

The people who produce the API are considered a critical partof the system and, as such, become part of the requirementsfor quality of APIs. To do their jobs effectively and to assurequality of the API, they must be properly trained andequipped. Qualified personnel must conduct the training;the equipment must be of proper design and function. Thesupervisors of people manufacturing APIs must also be prop-erly trained to do their jobs. Finally, there must be an ade-quate number of people to allow sufficient time to performthese responsibilities in a satisfactory manner.

E. The Quality Management Department

As in most any other manufacturing enterprise, there is a qual-ity control and=or a quality assurance department. Today,these departments are usually combined into a QM depart-ment. The role of the QM department has also advanced from‘‘check-test-decide’’ responsibility to being an equal partner

Quality Assurance and Control 237

Page 253: Active Pharmaceutical Ingredients

with manufacturing and engineering to manage and improvethe quality of the entire process and system.

For APIs and drug products, the QM department, throughits quality assurance arm, still has the responsibility vested init by regulations to release all products for use and eventuallyto the market. As a component of the system to produce APIs,the activities and responsibilities of the QM department arealso a component of product quality. Most cGMPs require thatthe QM department is responsible to review and approve pro-duction procedures, and any changes to them, most reports,procedures, and controls, deemed necessary to assure the qual-ity of the process and product.

Finally, the QM department must have adequate labora-tory facilities and properly trained and experienced people toeffectively carry out their responsibilities.

F. The Regulatory Authorities

Health authorities in every country regulate drug products.In most countries, these regulations also include APIs. ThesecGMP regulations require that a drug must meet all prede-fined quality specifications and be produced from a documen-ted validated process. Further, if the drug, or API, is notproduced and controlled according to the established process,then the drug is considered adulterated, and therefore not fitfor use or sale. The regulations address every aspect of drugproduct manufacture, and essentially require that the produ-cer has documented evidence of proof of control over anyaspect that might affect product quality.

The regulators were deliberate in their use of the word‘‘current’’ when the cGMPs were promulgated. This qualifierenables the agencies to continuously require that manufac-turers maintain their facilities and processes at the state ofthe art, thereby always assuring the public that drugproducts are as safe and effective as possible.

G. The Regulations

The production of APIs is regulated in most countries. TheICH-harmonized tripartite guideline Q7A entitled as Good

238 VanderZwan

Page 254: Active Pharmaceutical Ingredients

Manufacturing Practice Guide for APIs was recommended foradoption at Step 4 of the ICH process on the 10th of November2000. This document was adopted by the following agenciesdenoting its widespread acceptance:

� European Union (EU) adopted by CPMP, November2000, issued as CPMP=ICH=1935=00

� Japanese MHLW adopted November 2nd, 2001 MSBnotification NO. 1200

� United States FDA published in the Federal Register,Vol. 66, No 186, September 25th, 2001, pages 49028–49029.

The production process and all tests and controls must beapproved by the regulating government in which APIs will beused, and the facilities and systems in which they are pro-duced must meet the manufacturing standards set down bythe governing body. Thus, the quality of APIs is based ontwo components: meeting final quality specifications andbeing produced according to the regulated, approved processin a facility compliant with the appropriate manufacturingstandards. It is important to note that both criteria must bemet: final specifications and compliance to manufacturingstandards. These two components will be dealt withseparately in this chapter. It is also important to note thatthe approach toward quality described in this chapter shouldapply to any API regardless of the country in which it will beused or sold, or whether or not it will be a regulated item.The approach to quality, described in this chapter, is basedon sound scientific principles, good QM principles, andapplies to any API. In fact, these principles apply to themanufacture of any chemical that requires a high assuranceof quality.

This chapter will deal with the chemical synthesis ofAPIs. However, all the principles and regulations also applyto other means of preparation, such as fermentation routesor extraction from natural sources.

Finally, since it is assumed throughout this chapter thatthe API will be subject to regulatory requirements, reference

Quality Assurance and Control 239

Page 255: Active Pharmaceutical Ingredients

will be made to the regulations. If the reader is dealing withan unregulated item, such reference may be ignored, butthe scientific principles on which the regulation is basedshould be seriously considered.

II. DEFINING AND ASSURING THE QUALITYOF THE ACTIVE PHARMACEUTICALINGREDIENT

This section of the chapter addresses how to:

� define the necessary quality attributes� test for them,� design them into the process, and� validate the process to assure consistent production.

As APIs are regulated articles, their quality is determinednot only by satisfactory test results, but also the assurance thatthe process was conducted according to a validated process.

A. Defining the API Quality

The API must have its final chemical purity and impurity andits final physical attributes specified; some articles alsorequire microbiological analyses to be determined, dependingon the final dosage form and the manufacturing processinvolved. These attributes are established to assure an APIwill perform satisfactorily in the pharmaceutical manufactur-ing process and will result in a final dosage form; i.e., the drugproduct that will meet its initial release specifications andfinal stability requirements. The chemical purity minimumis usually set at 98% to assure proper dosing in the drugproduct and to assure a minimal amount of impurities. Thephysical parameters should be established with knowledgeof the pharmaceutical process and the ultimate final dosageform. Other attributes usually include color of the solid formand=or a solution, melting point, specific rotation if opticallyactive, crystal morphology, and so forth. A list of typicalAPI specifications is provided in Appendix A along with therationale for each one.

240 VanderZwan

Page 256: Active Pharmaceutical Ingredients

When setting API physical attribute specifications,the most important aspect to consider is its use in the pharma-ceutical process; namely, whether it will be wetted for granu-lation, dissolved for solution, dry blended, and so on, and thetype of drug product to be made: tablets, capsules, solutions,sterile or non sterile, or other. It is also important to knowhow the drug product will be used by the patient; for example,if it will be used as a powder blended with other excipients,careful consideration should be given to rate of dissolutionand the eventual color of solution (for aesthetic reasons) whendissolved by the patient (or healthcare giver) prior to use. Forthis reason, final API specifications are always defined withthe cooperation of the pharmaceutical development area.The quality assurance function approves final API qualitystandards, taking into consideration all requirements: processrelated, governmental, and customer.

B. Testing the API for Its Defined Attributes

Each quality attribute required of the API must have a soundand proven test procedure. In regulatory compliance terms,this means the test must be validated; that is, to have docu-mented proof that it performs reliably, is indicative of theattribute under question, and is not biased by interferingcomponents. There are eight specific components of a vali-dated test, and for an excellent treatise on this, the readeris referred to the current USP or the ICH guidance on analy-tical test validation. Most regulatory authorities require a testfor all significant API quality attributes on each lot produced.In nearly all cases, the pharmaceutical manufacturer requiresa certificate of analysis (C of A( documenting the resultsobtained on each lot, as well as a statement from the qualityoffice that the batch met its established quality criteria.

C. Designing Quality into the Process

As described above, the pharmaceutical manufacturing pro-cess and end use of the drug product dosage form are the basisfor establishing the limits of chemical purity and physical

Quality Assurance and Control 241

Page 257: Active Pharmaceutical Ingredients

attributes. Having predefined these attributes, the syntheticchemist and chemical engineer have the task of designingquality into the process; thereby assuring every lot will meetits criteria. This is perhaps the most significant aspect of che-mical process validation and a cornerstone of most regulatoryrequirements for quality assurance. After the chemical pro-cess is developed, a technical document, which explains howand why certain reagents, steps, controls, etc. were chosenin order to build quality into the product, should be prepared.When themanufacturing team takes on the commercial imple-mentation of the process, and goes through the formal manu-facturing validation process, they should rely heavily on thistechnical document to prove the quality of the final API. Asstated in the introduction, quality is designed into the processnot for regulatory purposes, but because it makes good manu-facturing and business sense to do so. Manufacturers want aprocess that safely and reliably delivers high yield and qualityfor economic and environmental reasons.

One should begin the approach to designing quality intothe API, with the concept of designing a perfect system. Keepin mind that all the safety, environmental, and economic rea-sons for developing a perfect chemical synthesis are preciselyconsistent with the goal of designing quality into the process,and very well serve all regulatory process validation and con-trol requirements. If one imagines a perfect process, there willbe no toxic emissions about which to be concerned, no safetyconcerns or need for special safety controls, and the yield ofeach step will be 100% of the desired intermediate, stereo iso-mer, and end product. Such a process would be free of anyimpurities and would assay for 100% purity. The nextchallenge is to design the synthesis so that each step can beprecisely controlled to always provide the same end result.The design work requires a complete understanding of thechemical reactions in the synthetic process under develop-ment. Then a clever design can be developed to eliminateany undesirable side reactions. In some instances, this canbe achieved by sophisticated use of functional group protectingagents, and in other instances by changing the sequence offunctional group introduction onto the end product building

242 VanderZwan

Page 258: Active Pharmaceutical Ingredients

block and sometimes by simple careful control over reactionparameters. Once the process has been perfectly designed,developed, and controlled, the last concern is over the controlof quality and reliability of the raw materials, proper function-ing of equipment, and error-free operations by personnel. Withthe vision of a perfect system in mind, one can imagine how theAPI quality would be perfect and consistent.

D. Validation of the Process

This aspect of the regulations is perfectly aligned withbusiness interests. The regulations require that a chemicalmanufacturing process be validated, which the author person-ally defines as proof of knowledge of control.

While the term ‘‘validation’’ has various definitions inseveral different regulations (cGMPs), all essentially meanor imply ‘‘proof of knowledge of control.’’ In essence, the vali-dation of the process is the description of the process afterall development work is completed, with the elaboration ofthe proof of synthetic pathway, controls over process condi-tions, and finally, sound analytical proof of quality fromsamples obtained during actual manufacturing campaignsin the plant. Critical process parameters such as time, tem-perature, and mixing conditions should be defined, con-trolled, and monitored. The kinetics of the syntheticpathway is documented in a process manual. The establish-ment of a process manual for each API is the foundation ofprocess validation. In this manual, one describes proof ofthe knowledge of the process and the controls necessary forconsistent results. Hence, the scientific design process tobuild the perfect process requires full knowledge of thechemistry of the process. That knowledge is described inthe chemical pathway from raw materials to the final API.The scientific evidence, such as intermediate structure eluci-dation, spectrographic analysis (IR, Near IR, mass spec, UV,NMR, C13NMR, etc.), and the proposed chemical mechanismfor each transformation, serves as the proof of that knowl-edge. Finally, during the course of the process development,full knowledge is gained concerning those parameters and

Quality Assurance and Control 243

Page 259: Active Pharmaceutical Ingredients

conditions that affect the kinetics, yield, and purity of eachstep. Experiments to optimize each step for purity and yieldlead the process engineer to describe the necessary controlsand conditions. These controls are described in a processmanual and are used in the scale-up work and ultimatefull-scale operation in the chemical plant.

E. Reality

We realize that the perfect synthetic process will, in alllikelihood, be too elusive. Eventually, we must make thedecision to focus our resources on the best process availableafter thorough development work yields a sound and reliableprocess.Eachsynthetic challenge represents reality of thebusi-ness of APImanufacturing, and so at some point, the feasibilityof further studies vs. commercializing what has been achievedto datemust be evaluated on a risk (loosing precious time in themarket) to reward (achieving a superior process) basis. It is suf-ficient to say here that to ensure quality of the final API, thedevelopment of the process provides the necessary informationto design in-process controls needed to monitor the progress ofeach step. These controls are the chemical and physical moni-tors that inform the operator that the synthesis is proceedingaccording to the original design. They are used also to informthe operator when the reaction is complete and when the nextstep may occur. In many cases, especially when the process iswell defined and designed, including the quality of startingmaterials and reagents, a good control is simply the use of time,based on a knowledge of the kinetics of the reaction.

In-process controls should always be ‘‘in the process,’’that is, ‘‘on-line,’’ and not requiring a sample to be withdrawnand sent to a laboratory for testing and evaluation. Undersome conditions, it may be necessary to take samples, but thisshould be avoided whenever practical.

In-process controls are probes, or monitors, inserted intothe reaction vessel, or the gauges that measure and recordpressure and temperature of vapors above the reactionmedium. The attributes that are measured include a widevariety relevant to the specific chemistry taking place. Prop-erly established tests, for example, infrared or ultraviolet

244 VanderZwan

Page 260: Active Pharmaceutical Ingredients

analysis, can predict the end point of a reaction by followingthe disappearance of a functional group on a reagent, or theformation of one on the molecule being produced. Monitoringthe presence of any side products, such as water or gases, willsignal the end point of the reaction when their theoreticalyield is obtained.

III. THE REGULATIONS FOR QUALITY

A. Introduction: The Emergence of SpecificRegulations for APIs

The active ingredient of a pharmaceutical product must meettwo distinct sets of criteria before it can be used for producinga drug product suitable for sale in most countries around theworld. One set of criteria is the product specifications,addressed in Part II. The other set is the assurance that theproduct is produced according to cGMP, that is, the cGMPsprevalent in the regulated market in which the drug productwill be sold. Most countries have approved and enforced reg-ulations for drug products; there were few with specific regu-lations for the APIs used therein. With the development andsubsequent adoption of the ICH Q7A Guide, by the EU,MHLW and US FDA, a consistent approach to cGMPs forthe manufacture of APIs is now achievable.

The section is written as if the regulations are in forcethroughout the world. This position is valid given the adop-tion of the ICH Q7A Guide.

One final introductory comment before beginning areview of the ICH guidance: when describing the ‘‘cGMPs,’’they are always prefaced by the adjective ‘‘current’’. Q7Aacknowledges the equivalence of the terms ‘‘cGMPs’’ and‘‘good manufacturing practices’’. The equivalence of the termsis deliberate. It requires that manufacturers continuouslyapply the current state of technology and practices whendeveloping new drugs. In certain special cases, manu-facturers will also be compelled to apply the new technologyto older APIs and the processes, facilities, etc., whenever suchapplication will play a significant role in assuring, or

Quality Assurance and Control 245

Page 261: Active Pharmaceutical Ingredients

advancing, the end product quality. Furthermore, since the‘‘current’’ is part of the guidance, manufacturers need to beaware of such advances and make the necessary changes totheir systems and facilities to remain compliant. Hence, theguidance can be thought of as always being updated withoutreturn to the regulating bodies for approval.

This part of the chapter follows the format of the ICHGuide as finalized by ICH in November 2000 (available fromwww.ich.org—see ‘‘Quality,’’ then ‘‘Q7A’’; or www.fda.gov).However, it is not intended that this will represent a sum-mary of the guidance. Instead, this text offers practicalinsight into the reasons and meanings of certain aspects ofthe requirements. Review and reference should that be neces-sary. The reason for selecting the ICH guide is due to itswidespread adoption, its comprehensive approach and itshigh quality as a reference document.

The ICH guidance is laid out in the following formatto demonstrate the scope and extent of their influence onthe entire manufacturing process (note we are followingthe ICH numbering format), each of these ICH sections arediscussed below:

1. Introduction2. QM3. Personnel4. Buildings and facilities5. Process equipment6. Documents and records7. Materials management8. Production and in-process controls9. Packaging & identification labeling of APIs and

intermediates10. Storage & distribution11. Laboratory controls12. Validation13. Change control14. Rejection and reuse of materials15. Complaints and recalls16. Contract manufacturers (including laboratories)

246 VanderZwan

Page 262: Active Pharmaceutical Ingredients

17. Agents, brokers, traders, distributors, repacka-gers and relabelers

18. Specific guidance for APIs manufactured by cellculture=fermentation

19. APIs for use in clinical trials20. Glossary

(Note:Sections17–20arenotdiscussedwithin this chapter).To assure the manufacture of an API meets the require-

ments described in the ICH guide is, indeed, a significanttask. It requires that all the requirements are understoodby all appropriate people dealing with the manufacturingprocess, that their understanding is proven through docu-mented training records, and there are effective systemsand procedures in place to assure that all appropriate steps,controls, tests, etc. are conducted, as described, in the pro-duct’s New Drug Application, in the firm’s Drug Master File,and in the documented standard operating procedures of amanufacturing facility.

Let us begin with an analysis of the ICH Q7A Guidelines,section by section.

1. ICH Q7A Section I: ‘‘Introduction’’

This section describes the scope and application of the guide-lines. It provides guidance as to when cGMPs should beapplied to the manufacturing process. The grid from theQ7A Introduction (Fig. 1) demonstrations GMP applications.It shows the various types of API manufacturing technologies,for example, from ‘‘chemical manufacturing’’ on the top left ofthe grid through to ‘‘classical fermentation’’ on the bottomleft. For each technology, moving left to right, the likely pro-cessing steps that might be used are mentioned. As the pro-cess moves closer to the final steps, the degree of cGMPrequirements increases. At some logical point, the heads ofmanufacturing and quality decide on the ‘‘starting materials’’that will reliably produce the API. These starting materialsmust be very well characterized, and always be tested for con-formance to predefined attributes before use in the API pro-cess. Admittedly, which chemicals are defined as ‘‘starting

Quality Assurance and Control 247

Page 263: Active Pharmaceutical Ingredients

materials’’ can be debated. Logically, if a reagent will becomepart of the final molecule, it is a very good candidate to bechosen as one of the ‘‘starting materials.’’ From this point for-ward, the cGMPs applied to the process increase in theirstringency. Final packaging of the APIs will be very well pre-scribed in both the environmental requirements and in thelabeling controls applied.

Figure 1

248 VanderZwan

Page 264: Active Pharmaceutical Ingredients

a. Compliance Requirements

Defining the point at which cGMPs first come into effectfor the API (or intermediate) being produced is the essence ofthis section. From that point forward, the strategy and therequirements for consistently producing APIs in conformancewith cGMPs are developed.

2. ICH Q7A Section 2: ‘‘Quality Management’’

Beyond the principle that quality is the responsibility of allpersonnel, there are specific responsibilities, which must becarried out by the quality unit. The challenge is to achieve abalance between executing the necessary activities and notallowing the abdication of other departments toward quality.The quality unit most often becomes a focal point for all qual-ity related matters, functioning as a technical consultant inquality and compliance. It is imperative that the quality unitbe independent of the manufacturing operations in order toachieve an objective perspective. Some of the basic qualityresponsibilities include review and approval of documents(specifications, test methods), written SOPs for all depart-ments, records (batch records and log books), deviations andtheir investigations=resolutions, and finally the release ofthe product to market.

Internal auditing (also termed self-assessment and self-inspection) is also a cornerstone of QM. Knowledge of theplant and its systems should be used to determine the annualschedule for conducting audits. Once executed, the infor-mation contained in an internal audit is useful not only forthe department being audited to improve their operations,but also for the plant on a larger scale to know about potentialquality issues in advance. Senior management must be madeaware of the issues found in internal audits since they areresponsible for setting the strategy for the plant and can allo-cate resources to correct any deficiencies.

In addition to internal audits, which are carried out onmanufacturing processes, a review of the product as it ismanufactured, and its ability to meet specifications bothinitially and over time, can yield information for improving

Quality Assurance and Control 249

Page 265: Active Pharmaceutical Ingredients

processes and the product itself. These reviews are typicallycaptured in an annual report known as the product qualityreview, annual product report or product quality history. Eachplant defines the scope of the report, however but minimallythe results of all tests (at the time of release and when testedsubsequently), the review of deviations and difficulties in itsmanufacture, complaints received from customers, and espe-cially looking for trends that may not be apparent on a day-to-day basis are the foundation of any product review.

a. Compliance Requirements

A well-defined internal auditing program, comprehen-sive in scope with a good communication plan, can helpidentify and prevent systemic problems in the operation.The internal auditors must not only have a thorough knowl-edge of the operations and the regulations, but also the bestmeans of achieving conformance without creating unneces-sary bureaucracy. Combined with annual product qualityreviews, opportunities for quality improvement should beidentified both for the products and the processes.

3. ICH Q7A Section 3: ‘‘Personnel’’

No matter what API is made, there are always peopleinvolved in the process. It has been said by numerous CEOs,‘‘people are our greatest resource.’’ Each manufacturingoperation needs the right level of personnel both in terms ofnumber and qualification for each job in the operation. Thispresupposes that each job has a well-defined job descriptioncomplete with training requirements and the demonstrationof the proficiency of the necessary activities. While this hasbeen common in the laboratories, companies are now expand-ing the concept of training and qualification to the informalprograms common in our industry. The use of ‘‘mentors’’exists in most manufacturing operations but few ask, ‘‘howare these mentors chosen, what are the skills that make oneperson an excellent mentor and the other a poor one, whattraining is needed to make the mentoring effective andefficient?’’ The recognition of mentoring as a training activitywill increase the knowledge base resident in your work force.

250 VanderZwan

Page 266: Active Pharmaceutical Ingredients

Since people are involved in the manufacture of APIs, theirhygiene becomes important. The firm must provide adequatetoilet, cafeteria, and changing=locker facilities. This achievesnot only protection of the personnel, but also of the product.

It is common practice to employ consultants or contrac-tors for limited times on short-term specific projects. Consul-tants or contractors must follow the same requirements forpersonnel hygiene and must have the training necessary toperform their duties.

a. Compliance Requirements

All personnel involved in the manufacture of APIs andintermediates must have the necessary training, education,and skills to perform their activities in a consistent manner.This must be documented not only for regular employeesbut also for contractors=consultants as well. Proper hygieneand sanitation protecting the product and the personnel mustbe part of normal operating conditions at the site.

4. ICH Q7A Section 4: ‘‘Buildings and Facilities’’

In planning a facility, there are always drawings of the facil-ity; the equipment, the utilities, the material flow, and per-sonnel flow. This is necessary to assure that in the design ofa facility, adequate space is provided for the material and per-sonnel to flow smoothly, and for the prevention of mix-ups andcontamination.

Utilities can have direct impact on the quality of theAPI. The utility can either be required to assure consistentenvironmental parameters, as evidenced by heating, ventila-tion, and air conditioning (HVAC) or by providing materialsthat come in direct contact with the product (compressedgases). Inconsistency in either of these functions can resultin inconsistent quality of the product and=or process. As such,all utilities having a direct impact must be qualified throughrigorous testing to show it will consistently perform asexpected. In addition, these utilities have to be monitored toassure they are continuing to function as necessary.

Water canbeused for a variety of purposeswithin theman-ufacturing of APIs. Because of the many differing uses, such as

Quality Assurance and Control 251

Page 267: Active Pharmaceutical Ingredients

a cleaningmaterial or a rawmaterial in themanufacturing pro-cess, how the water is used will determine the specifications tobe developed. Beyond how the water is used, the product it isused in will also determine the specifications. At a minimum,process water must meet World Health Organization’s (WHO)guideline for drinking (potable) water quality. If this water isthen treated to yield a water of a predefined quality, thatprocess must be validated and the water produced monitored.This can include specifications for themicrobial content or endo-toxins in the water for an API to be used in a sterile product.

Containment considerations must also be built intothe design of the facility and their utilities in order to pro-tect the workers from adverse exposure to the API, or itsintermediates. Certain APIs are highly toxic or potentiallydeleterious materials, such as penicillin and cephalosporins.The side effects from these drugs can be life threatening. Itmay be necessary to develop separate and dedicated facilitiesfor the manufacture of these types of compounds.

Lighting should be sufficient for all personnel to performtheir activities without eyestrain. Certain APIs may need tobe protected from light. In those cases, the lighting may haveto be designed taking into account the product’s requirementsand the personnel’s needs.

Every operation will generate waste. In the case of themanufacturing of APIs, this waste can cause contaminationthroughout the manufacturing facility if not removed in asanitary and safe manner.

How a facility is maintained is one of the first indicationsof the quality culture at the site. A high-quality API requiresa facility that is clean, free of pests and has utilities that func-tion reliably. The cleaning agents or other materials used tomaintain the facility of equipment must be known not to con-taminate the product in its usage. Reliability of the utilitiescan be assured by a well-defined and executed preventivemaintenance program.

a. Compliance Requirements

Engineering drawings detailing the facilities layout,equipment location, room usage, material flow, personnel

252 VanderZwan

Page 268: Active Pharmaceutical Ingredients

flow, and utilities must be maintained to reflect accurately thebuilding and facility. Procedures for the maintenance andmonitoring of all the utilities that can impact product qualitymust be written and approved. Utilities producing materialsthat are used in the actual manufacturing operations musthave specifications for those materials. Safety of the person-nel and the prevention of the product contamination mustbe thought of in advance and captured in standard operatingprocedures specifying sanitation, containment, and refusedisposal practices. The maintenance of the facility, the build-ing, and the utilities must be defined in operating proceduresusing materials that will not adversely affect the product.

5. ICH Q7A Section 5: ‘‘Process Equipment’’

For each manufacturing operation, there will be equipmenteither fixed or mobile to be used in the process. Sometimesthe equipment may be placed outside of the building itself.Equipment can be either closed (preferable) or open. If theequipment is not closed, there must be special attention toprevent contamination of the product. In either case, theequipment must be constructed of the right materials toassure that it can be easily cleaned and maintained. It mustprotect the product it is manufacturing and not affect thequality of the product. Identification of the equipment (andprocessing lines) is necessary to assure traceability of the pro-duct to the equipment used. Not all equipment will need to beidentified; each facility must define which pieces of equipmentare considered major in their own manufacturing processes.The use of lubricants or other manufacturing aides is oftennecessary, however, when used, these materials must notcontact the API or alter the quality of the API.

A clean, well-maintained building is only half of the pic-ture. A clean facility with broken or rusty equipment is just asindicative of a poor quality culture as is the reverse. Theequipment in the building must be maintained in a good stateof repair and cleanliness. The expectation must be that onceprocessing starts, all the necessary equipment will functionproperly.

Quality Assurance and Control 253

Page 269: Active Pharmaceutical Ingredients

Cleaning materials must be chosen not only to clean theequipment, but also to leave no residues, which may affectthe quality of subsequent batches. How long a piece of equip-ment remains ‘‘clean’’ after cleaning is completed is animportant factor to be considered in designing the cleaningprogram. Also part of a comprehensive cleaning program iswhether reduced cleaning can be used for campaign manu-facturing (subsequent batches are produced of the identicalAPI).

Manufacturing equipment often provides data, informa-tion and output, which can be used to determine the accept-ability of the processes and=or the product itself. We mustbe able to rely on this data as correct and accurate. Therefore,the analytical components of this equipment must becalibrated using standards whose authenticity is assured.The time in between calibrations will be based on the reliabil-ity of the equipment and the criticality of the data. When cali-bration requirements are not met, decisions based on the datamay be erroneous and must be re-evaluated. Instruments,which do not meet calibration requirements, should not beused and there must be an investigation to assess the impacton batches produced in that equipment.

Computerized systems are specialized pieces of equip-ment which must meet all the requirements as other piecesof equipment. A logical application of the cGMPs should beapplied in conjunction with special requirements such as211 CFR Part 11. Validation is required, however, the depthand scope of the validation is dependent of the computerizedapplication. The degree of validation may be dependent onthe source of the computer system: commercially availablesoftware requires little validation, while software developedfor a specific manufacturing step will require extensive vali-dation. Once validated, the computer system must be main-tained in a state of control. After all, it is still a piece ofequipment, albeit highly specialized.

a. Compliance Requirements

Procedures for the cleaning, maintenance, and operationof each piece of major equipment must be developed and

254 VanderZwan

Page 270: Active Pharmaceutical Ingredients

approved by the quality organization. There also needs to beengineering drawings of the equipment along with the main-tenance of a revision history of these drawings. Many firmssee engineering drawings analogous to documents andrequire quality approval of changes.

6. ICH Q7A Section 6: ‘‘Documents andRecords’’

In a sense, our industry prepares two products: the API thatgoes out the door and the paperwork which records how thisbatch was made. The paperwork consists of documents suchas procedures, specifications, methods, and manufacturinginstructions. These documents are prepared and revised bya formal change control process; the quality unit must reviewand approve them. The issuance of these documents to theoperators, technicians, and other persons using these docu-ments must be done in a controlled manner, assuring theyhave the most recent version of the approved document. Typi-cally, the quality unit has a documentation center which willstore the superseded documents and the approved mastercopies. The storage of documents must be defined in a recordsretention policy consistent with cGMP requirements (at least1 year after the expiry of the batch or for APIs with retestdates, 3 years after the complete distribution of the batch)and any legal requirements of the firm.

The data detailing the actual manufacturing conditions(temperatures, lot numbers of starting materials, etc.) isrecorded on a batch record. Data detailing equipment usageand conditions are similarly considered to be a cGMP record.Changes to cGMP records must show the person making thechange and the date the change was made. Many firms alsoannotate the correction with the reason for the correction.

Only accepted raw materials may be used in the produc-tion of an API. This sets into motion a requirement for properdocumentation surrounding the receipt, testing, acceptance,and release to manufacturing. Imagine that a raw materialis found to be defective after it has been used in a product.

Quality Assurance and Control 255

Page 271: Active Pharmaceutical Ingredients

The firm would have to trace (using the lot number of the sup-plier) to determine which batches this defective raw materialhas been used in. The records then must include the suppli-er’s lot number, a unique lot number assigned to the materi-als, date of receipt of the material, and records of the testingand release of the material.

Manufacturing Instructions are documents detailing theprocess and the controls necessary to produce uniform qualitybatches, time and again. In a sense, the manufacturinginstructions are the recipes for making the API.

Specific requirements for these records include:

� Name of the API and any unique identifying code.� List of raw materials and the quantity or ratio of

materials to be used. Specific calculations must beincluded as part of the specific batch record.

� List of equipment to be used.� Specific production instructions in the correct

sequence with all necessary control parameters, timelimits, yield ranges and in-process control testingand specifications.

� Instructions for storage.

There should be a master batch record document fromwhich each individual batch record is generated. This insuresthat each lot made is manufactured following the same recipe.When the lot specific batch record is issued, there must be acheck to assure that the issued batch record is specific for asingle unique batch number and the instructions are identicalto the master batch record document. The lot specific recordwill have the signature of those individuals who performedcritical processing steps in the manufacturing process andthose who performed any in-process testing.

Eventually, irregularities will occur. Chemical manu-facturing processes are a complex interaction of reagents,solvents, machines, and people. Machines can break downand people can make mistakes. In-process materials, or pro-ducts do not always meet specifications. Each firm must havea system in place to identify and assess the impact of thesedeviations on product quality and process robustness. When

256 VanderZwan

Page 272: Active Pharmaceutical Ingredients

deviations occur, they must be addressed as part of the batchrecord. Prior to the release of the batch, the quality unit mustapprove the deviations and how they have been resolved.

As the batch record details the executed processing steps,there are records for the laboratory testing for each batch.These requirements are completely consistent with thecGMPs for laboratory records for finished pharmaceuticals.Samples are taken to determine the acceptability of a mate-rial; the traceability of a sample to the portion of the lot whereit came from must be known and recorded. We must knowdefinitely what tests were performed on the sample (testmethod including revision date), what equipment was usedto generate the data, who performed the test, who reviewedthe data (a second person must review the data) in additionto the acceptability of the test results themselves.

When the manufacturing and the testing activities arecompleted, a review of the critical process steps and the non-critical process steps must be performed. The quality unitmust review the critical process steps; the noncritical processsteps can be reviewed by a different unit following proceduresthat have been approved by the quality unit.

a. Compliance Requirements

Any aspect of producing an API, from the receipt of allincoming materials from outside vendors right through tothe last distribution of released material, must be appropri-ately recorded. Systems must be in place so that each specificlot of intermediate or API is reviewed and approved by QCbefore it is released. Effective systems need to be in place toboth detect an unexpected result, and then to investigate it.Batch production records, or a sound sampling thereof, as wellas all other quality related records such as stability data com-plaints and so forth should be reviewed at least annually toensure that in-process controls, procedures, and final productspecifications are adequate and tightened where appropriate.

7. ICH Q7A Section 7: ‘‘Materials Management’’

There must be a comprehensive system with procedures defin-ing the receipt, identification, quarantine, storage, handling,

Quality Assurance and Control 257

Page 273: Active Pharmaceutical Ingredients

sampling, testing, and disposition of materials. There must bepredetermined specifications for materials purchased from asupplier, which has been approved by the quality unit. Thispresupposes that there is a system for approving suppliersand the materials they supply. Critical raw materials mustbe identified and changes to any specifications of critical rawmaterials must be handled under change control.

A manufacturing plant will receive several differenttypes of incoming materials, some require the strictest degreeof controls (raw materials and intermediates, for example)and others can be handled with little more than good account-ing practices (office supplies, etc.). For each material to beused in the manufacturing process, there must be a uniquelot number assigned to the goods to assure complete traceabil-ity to the supplier and the shipment. Shipping conditions canaffect materials even during a short time; shipments of thesame supplier lot number may require a different receivinglot number. Upon receipt, the containers should be checkedfor conformance to the labeling and be free from tamperingor damage, which may cause contamination of the API. Beforeany material is used, the quality unit must formally releasethe material.

The use of nondedicated tankers requires an additionallevel of assurance that any potential contamination is pre-vented. This can be assured by audits of the suppliers and=or a certificate of cleaning supplied by the supplier and=ortesting for impurities by the receiving firm.

Many firms rely on a C of A supplied by the supplier inlieu of actually performing required testing. This becomes amore proactive means of assuring the quality of the material.Acceptance of a C of A is possible after a partnership is estab-lished with the supplier through a formal qualification andevaluation of the supplier’s capabilities and reliability. Typi-cally the process involves an initial questionnaire, followed byan audit by trained auditors and purchasing representatives.This helps determine if the supplier is qualified to produce thematerial consistently and in accordancewith the firm’s expecta-tions. Thematerial thenneeds tobeapproved foruse in theman-ufacturing process; this is typically done with three distinct

258 VanderZwan

Page 274: Active Pharmaceutical Ingredients

batches produced by the supplier. The goal is to determine if thesupplier’s material performs reliably in the firm’s manufactur-ing process. The entire process of qualifying the vendor andthe material they produce must be repeated on a periodic basis.For hazardous or highly toxic raw materials, full acceptance ofthe C of A may be warranted pending a documented rationale.

It would be easy if all materials were received as a singlelot, in a single container; however, it is often the case that thevendor will use multiple containers—and sometime differentlots, typically to facilitate his own handling and shippingefforts. Each container must be inspected at the time of receipt;however, the contents of each container need not be sampledand tested to determine the material’s acceptability. The useof statistical sampling plans applied to each lot separatelycan help reduce the burden of sampling and testing while stillyielding a result representative of the batch as a whole. Thesesampling plans must take into account the number of contain-ers received and the criticality of the material.

While these materials are held, either prior to release ininventory or in the manufacturing process itself, they must beheld and handled so as not to contaminate the material itself,or other materials stored in the area. For example, heat sen-sitive materials may need to be stored in a cool, controlled(i.e., data-recorded) location. It is quite common in APImanufacturing to store materials outdoors. This can beaccomplished for specific materials as long as the require-ments stated above are met. When brought into the manu-facturing environment, the containers may require anadditional cleaning.

If a material is determined to be unfit for use andrejected, special storage and handling requirements must bemet. It must be stored in such a manner that it cannot be usedinadvertently in the manufacturing process. Many firms havedesignated locked cages to store rejected materials.

a. Compliance Requirements

The receipt, storage, and handling of materials must beperformed in such a manner that there is complete traceabil-ity of the material to the supplier. Materials may be received

Quality Assurance and Control 259

Page 275: Active Pharmaceutical Ingredients

against a C of A supplied by a qualified supplier in lieu of fulltesting. For this to happen there must be a formal supplierqualification process. For materials that are sampled andtested for release, there must be predetermined statisticallyvalid sampling plans and acceptance criteria. Materials mustbe segregated or otherwise stored to prevent their use, untilthe formal release by the quality unit is granted. The materi-als must be stored in such a manner as to not compromisetheir quality attributes. Any rejected materials must not beused and must be stored in a separate area to prevent suchan error. Rejected material should be returned to the vendor,or otherwise properly destroyed.

8. ICH Q7A Section 8: ‘‘Production andIn-Process Controls’’

Earlier, the need for a master batch record document was dis-cussed. This document describes the manufacturing instruc-tions necessary to consistently produce batches of APIs thatmeet predetermined specifications. There will also be ancillaryprocedures, which will define all the conditions and their con-trol parameters necessary to assure consistency from batch tobatch. Isolated materials should be labeled at each step in theprocess. This is true not only for the raw materials but also asin-process materials are generated and isolated, the material’sname, lot number, and its status should be clearly labeled. Asstated earlier, major pieces of equipment should be clearlylabeled with a unique identification number and its status(cleaned and ready to be used, to be cleaned, or in use). If a pro-cessing step is determined to be a critical processing step, itmay require witnessing of its completion by a second person,with the witnessing documented on the batch record.

Processing steps should have time limits, either a stepmust be completed within a certain time (mix for 2 hr) oran in-process material may be held for a specified amount oftime. Deviations from these time limits must be addressedwith a formal investigation as to the affect they may haveon the quality of the product. The results and conclusions ofthe investigation must be documented.

260 VanderZwan

Page 276: Active Pharmaceutical Ingredients

The manufacturing process should be reviewed for thosepoints where in-process testing can minimize variabilityin the process thus achieving greater consistency in the yieldand quality of the product. This review should ideally takeplace during development runs but information from vali-dation batches and annual product quality reviews can alsobe used. They can be valuable in determining which areasof the process or product should be more carefully monitoredor if process specifications should be changed. Types ofinprocess control tests include temperature of the process,pressure of equipment, the color of solutions, pH, loss ondrying, etc. In-process control testing and specificationsshould be defined in documents approved by the quality unit.

Each intermediate must meet its quality requirementsbefore further processing. Even if two or more batches ofintermediates will be blended prior to the next step, theymust each meet their respective quality requirements.

Blending is a process somewhat unique to the manu-facture of APIs. It is an accepted practice to blend batchesof the final API as long as each of the individual batchesmeets the predetermined specifications prior to the blending.After the blending is complete, there must be a final test toassure that the final blended lot is acceptable as well.

A sample of the final lot, blended or otherwise, should betaken and stored for future stability testing if necessary.

Campaign manufacturing is common in pharmaceuticaloperations. If successive batches are being made of the sameAPI, it is acceptable for residual materials to be carried fromone batch into another, as long as there is adequate control.Adequate control will need to be determined for each APIproduced but minimally there must be assurance that degra-dants, microbial contaminants, or other sources of contami-nation are not carried from one batch to another.

All operations must be conducted in such a manner thatcontamination is minimized. This is especially true after thepurification steps in the manufacturing process.

In addition to preventing contamination of the API,the safety of the operators must be addressed as well. Ifthe materials may be injurious to the operators and=or the

Quality Assurance and Control 261

Page 277: Active Pharmaceutical Ingredients

environment, how best to assure there is no harm to eithermust be addressed as part of the development of the manufac-turing process. It may be necessary, in some cases, for the pro-cess to be carried out using dedicated equipment or indedicated and controlled environments.

a. Compliance Requirements

Clearly, this section requires a large volume of docu-mentation, with the focus being proof that each batch hasbeen produced according to the original design. Each batchis individually monitored, and its manufacturing history isrecorded in a production batch record. In-process controlsshould be evaluated after adequate experience is gained infull-scale production. Test limits must be changed, i.e., tigh-tened, if justifiedbyhistorical results. TheQCunitmust reviewand approve all changes to production records, control proce-dures and test procedures and=or limits. Any deviation fromestablishedprocedures,whether planned or not,must be inves-tigated for cause, and documented for corrective action.

9. ICH Q7A Section 9: ‘‘Packagingand Identification Labeling of APIsand Intermediates’’

This section has four subsections, which state quite specificyet reasonable requirements for packaging materials, label-ing issuance and control, and packaging and labeling opera-tions of APIs. They are remarkable in their similarity to therequirements for drug products. They require tight controlover the receipt, testing, release, storage and use of contain-ers, and labels. Particular care must be taken to avoid mix-ups of labels, and separate storage areas should be providedfor all different labels. Further, access to the area should berestricted to only certain authorized personnel. The labelingoperations require the same assurance that the labeling facil-ities are separate from other activity and that they are ade-quately cleaned prior to use. All preparatory work must bedocumented in written procedures. Packaging and labeling

262 VanderZwan

Page 278: Active Pharmaceutical Ingredients

operations should be recorded in the manufacturing batchrecord for each lot of API.

The label must bear the usual descriptive informationabout the product and include its distinct lot number. Forintermediate or APIs with an expiry date, the date must beindicated on the label and on the C of A. For intermediatesor APIs with a retest date, the retest date should be indicatedon the label and=or C of A. Naturally, an effective systemmust be in place to assure no materials beyond their expiryperiod are used, and all those requiring retesting are com-pleted before use beyond the controlled time period.

As in other areas of API manufacture, contact surfacesmust not be reactive, absorptive, and so forth with the APIso as to alter its quality.

a. Compliance Requirements

Well-documented systems must be in place to handle thereceipt, testing, release, and use of containers and labels, simi-lar to those control procedures used for raw materials. Packa-ging and labeling operations must be conducted in separateareas to avoid contamination and mix-ups with other ongoingactivity. Inventory management of labels must be practiced,with accountability of all used and remaining labels kept up todate for each lot of labels. Expiry dates or retest dates must bebased on analytical evidence obtained under the intended sto-rage conditions, and each specific date for each lot must appearof all labels used to package each lot.

10. ICH Q7A Section 10: ‘‘Storage andDistribution’’

This brief section contains only two parts. It directs that thewarehousing procedures and distribution procedures mustbe written and, of course, be consistent with the intendedstorage conditions for which stability data exist for the mate-rial. Materials should be held in a quarantine condition untilreleased by the quality unit. This status control may be a phy-sical separation with appropriate labels or, ideally, throughthe use of electronic control systems. The distribution history

Quality Assurance and Control 263

Page 279: Active Pharmaceutical Ingredients

of each lot must be maintained for traceability in the event arecall is necessary.

a. Compliance Requirements

Have written procedures to describe the handling of thematerials. Ensure that a tight system is used to prevent use ofmaterials before release by QC. Material should be distribu-ted on first-in-first-out (FIFO) basis.

11. ICH Q7A Section 11: ‘‘Laboratory Controls’’

This section is described in greater detail in Part III of thischapter.

12. ICH Q7A Section 12: ‘‘Validation’’

This is a most important section of the cGMPs. As such, itwarrants its own chapter in this book. Here are describedthe essential compliance aspects of validation.

Within the ICH Guide, validation is further divided intothe areas of its disciplines:

� Validation policy� Validation documentation� Qualification (Equipment IQ=OQ)� Process validation� Periodic review of validated systems (Revalidation)� Cleaning validation� Validation of analytical methods

The validation policy is a high level document stating theapproach a firm will use toward validation. The validationapproach requires the development scientists and plant man-agement to identify the critical parameters=attributes duringthe development stage and use that knowledge in the valida-tion of the process.

For eachvalidation activity, there is a validationprotocol (astudy design) written and approved in advance of the executionofvalidationwork.Thequalityunitmust reviewandapprove theprotocol, as other affected departments. The protocol lists the

264 VanderZwan

Page 280: Active Pharmaceutical Ingredients

tests to be conductedalongwith theacceptance criteria; the testsare chosen to demonstrate the process is in a state of control.

Once the validation protocol is executed, the results ofthe tests are written into a formal report. Any deviations fromthe acceptance criteria must be addressed in the report, alongwith a conclusion about the impact on the consistency andreliability of the process.

Validation is a life cycle process, which has its roots inthe development area.

� Design qualification: verification that the proposeddesign is suitable for intended use.

� Installation qualification: verification that insta-llation complies with the approved design, manufac-turer’s recommendations, and=or user requirements.

� Operational qualification: verification that the equip-ment performs as intended throughout anticipatedoperating ranges.

� Performance qualification: verification that theequipment and=or process can perform according topreapproved specification consistently.

Just as there are phases in the validation lifecycle, thereare three distinct approaches a firm can take toward the vali-dation process.

1. Prospective validation is the preferred approach and isthe most common. If other approaches are used, the firmshould have documented rationale as to why they did notuse a prospective validation approach. A prospective valida-tion is a formal study that serves to prove the process willreliably yield an API to meet its predetermined quality attri-butes and all steps along the way are reliable in terms ofquality and yield. Validation can best be defined as proof ofknowledge of control. For new products, or changes to pro-cesses requiring a process validation, the number of runs mustbe commensurate with the complexity of the process, or thenature of the change under review. Three consecutive success-ful production batches are typically required; exceptionsshould be documented. Process validation must confirm theimpurity profile of the API.

Quality Assurance and Control 265

Page 281: Active Pharmaceutical Ingredients

2. Concurrent validation can be used where a small num-ber of API batches are made on an annual basis. Typically,there are three validation batches manufactured in a studyand all three are held pending the results. In the case wherethe time period between the production of the first batch andthird batch is extremely long, concurrent validation can beused. In this case, the first batch is released on its own merit,however, the process is not considered validated. Only whenthe full protocol requirements are met, both in terms of accep-tance criteria and in the number of batches are the processconsidered validated. The use of a concurrent validation isvery rare and only suitable under special circumstances.

3. Retrospective validation is very rare and must beused judiciously. This approach involves reviewing a largenumber of batches already produced at the plant to affirm therobustness and repeatability of a process. There are very speci-fic assumptions that must be met before retrospective valida-tion can even be considered. There must not have been anychanges made to the process during the review time period.The process must be a well-understood and -characterized pro-cess with defined in-process tests and controls. There must notbe any significant process failures or deviations during the timeperiod. The impurity profile for the product must be well estab-lished. Even when all these conditions are met, the decision touse retrospective validation must be a last resort and thejustification well documented with approval from the qualityunit.

Whatever approach is used for the validation, the goal isto gain the proof of knowledge of control of a process. A studyplan, called a protocol, is prepared describing the importantparameters that need to be controlled in order to assure theAPI will meet its quality parameters and expected yields.To determine those important parameters, data, results,and reports from the research department are used. Duringthe initial development of the process, the controlling para-meters should have been discovered, including effective work-ing ranges and targets for charge of components, raw mate-rials, and operating conditions of time, temperature,pressure, mixing rate, and so on. Analytical methods used

266 VanderZwan

Page 282: Active Pharmaceutical Ingredients

to evaluate each chemical and physical attribute are them-selves first validated. In this way, the data generated in thevalidation are known to be true and accurate.

The protocol defines how the study will be done (the pro-cess, equipment, critical steps, and parameters), and who isresponsible for its design, execution, analysis, and approval.The sampling activity is also well defined, describing the loca-tions to be sampled, the sampling devices to be used, thequantities required and time point during processing whenthey should be taken. The protocol defines all the importantprocess parameters to be studied and analyzed in order todemonstrate each significant step performs reliably in termsof quality and yield. The final approval is reserved for thequality function.

A successful validation study will demonstrate a reliableand robust process. To be able to reach a strong conclusion,there must be an adequate number of batches and tests to sta-tistically demonstrate reliability and robustness.

As part of the lifecycle approach to validation, periodicevaluations of the processes and products will determine theperiodicity of revalidation. Certain processes will require anannual revalidation. Revalidation also occurs when thereare significant changes to a process or piece of equipment,which would ‘‘void’’ the original validation.

a. Cleaning Validation

After manufacturing is completed, the equipment shouldbe cleaned and made ready for the next process. There needsto be a formal study, executed against a protocol, whichdemonstrates that the cleaning process used is effective toclean the equipment to a predetermined level of cleanliness.Cleaning validation is part of assuring that contaminationand cross contamination are prevented. The protocol mustinclude a description of the equipment to be cleaned, thematerials for cleaning to be used, and the cleaning process.The sampling equipment, locations, and procedures must bedefined. In addition to visual cleanliness, where analyticalmethods are used, these methods must be validated to appro-priate levels of detection. The limits of detection must be

Quality Assurance and Control 267

Page 283: Active Pharmaceutical Ingredients

based on sound scientific reasoning. Cleaning is not only per-formed to remove chemical contaminants but microbiologicalcontaminants as well. The removal of microbes and endotox-ins must be addressed in the protocol where appropriate.

The cleaning process must also be included when there isan evaluation as to the necessity of revalidations.

Analytical methods must also be validated. The approachused for method validation is consistent with the validation ofanalytical methods for drug products. See USP monograph onanalytical validation procedures.

b. Compliance Requirements

Assure the critical steps and intermediate quality attri-butes are defined and based on scientific rationale, usuallyfrom original research information. The person making thosedecisions must be identified in the protocol. Once the protocolis approved, it cannot be changed during the course of thestudy. While the documentation of validation studies is a reg-ulatory requirement, it serves the business aspects perfectlybecause it captures the intellectual property of the firm. Theprotocol should include ranges for operating parameters.These should come from research information. They neednot be tested or challenged during the validation study infull-scale equipment.

13. ICH Q7A Section 13: ‘‘Change Control’’

Having established a validated process, efforts must be imple-mented to assure that it stays in the validated state. Systemsneed to be implemented to evaluate both planned andunplanned changes to the process. This refers to any changein materials, conditions, equipment used, and site of manu-facture, scale, and so forth. All planned changes must bedescribed and evaluated before implemented. All concerneddepartments are involved in this analysis; the final reviewand approval is required of the quality unit.

Factors to be considered in evaluating the change shouldinclude any reasonable aspect of the API or its intermediatesthat may be affected. This must include attributes that are

268 VanderZwan

Page 284: Active Pharmaceutical Ingredients

not routinely tested, such as polymorphism, the emergence ofnew impurities, and the need for additional stability studies,for example. For this reason, chemical experts familiar withthe science must be consulted.

Finally, an analysis of the impact of the change on anyfiled regulatory documents is necessary, as well as informingthe pharmaceutical users of the API. Pharmaceutical manu-facturers might have additional quality and performance cri-teria, which are unknown to the API manufacturer, and thesecriteria need to be assessed relative to the process changeunder review.

The completely analyzed and studied process change isthen evaluated by the group of experts who designed thechange. The final review and approval are again required ofthe quality unit.

a. Compliance Requirements

An effective communication system needs to be in placeto ensure that the quality unit is informed and involved inplanned changes. Unplanned changes are to be discoveredthrough the periodic review of production records (see earliersection). Changes may be classified to their expected degree ofimpact, and studies can be modified accordingly. Scientificjudgment must always be used in evaluating the changes.Systems must be in place to ensure that material underchange review is not used for further processing untilapproved by the quality unit. The decisions about what toevaluate must be documented, as well as why no additionalstudies are deemed necessary (for example, why polymorph-ism will not be affected by the change).

14. ICH Q7A Section 14: ‘‘Rejection andRe-Use of Materials’’

This section describes the requirements for rejection of mate-rials, reprocessing and reworking, recovery and recycling ofsolvents in the process, and customer returns of materials.

If specifications are not met and=or if the material is notmanufactured in accordance with cGMPs, these materials

Quality Assurance and Control 269

Page 285: Active Pharmaceutical Ingredients

must be set aside, and quarantined until disposition. The dis-position can be rejection, reworking, or reprocessing thematerial. Material is used here to denote incoming materials,intermediates and=or finished product (APIs). If reworking orreprocessing is determined to be in accordance with regula-tory controls, the actual reworking or reprocessing must beconducted and recorded in a manner identical to that of theoriginal manufacturing steps.

The manufacturing of APIs can be distinguished fromfinished pharmaceuticals in that reprocessing is a far moreaccepted practice for APIs. ‘‘Reprocessing’’ is defined as thereturn of an intermediate or an API back into the processand repeating a part of the manufacturing step. Types ofreprocessing include either physical reprocessing, for examplethe repetition of a drying step, or extending a chemical step.However, if reprocessing is used routinely for any given step,at some point it becomes the normal process. At that point thereprocessing step(s) should be incorporated into the manufac-turing process and batch documentation, not as a reproces-sing step, but rather as a routine part of the process.

Reprocessing by chemical means involves the repeatingof a chemical reaction. This is rarely appropriate since inrepeating a chemical step, new impurities could be produced.A batch requiring chemical reprocessing should first be con-sidered for destruction before salvaging through reprocessingoccurs.

Reprocessing by physical means involves the repetition ofa step such as a recrystallization or remilling already routi-nely performed in the validated process. If the routine processdoes not include such a step, then it is not reprocessing butrather reworking. The actions to be taken in a reprocessingmust be documented with a documented rationale for thereprocessing. Reprocessed materials must be evaluated todetermine if additional testing is warranted.

‘‘Reworking’’ is distinguished from reprocessing in thatreworking is the use of a new step, or steps, not part of theroutine process. While reprocessing does not require a newand separate process validation, reworking must have itsown validation. Reworking requires the approval of the

270 VanderZwan

Page 286: Active Pharmaceutical Ingredients

quality assurance unit. Reworking often requires a notifica-tion to the governing regulatory authorities, and typicallyrequires their approval of the change before putting it intouse.

Recovery and recycling of reactants, intermediates, orAPI’s, in order to be used again, are considered acceptable.However, the use of these materials must be done using vali-dated and documented procedures. If a recovered solvent is tobe used in a different process (e.g., to produce a different API),there must be adequate validation and documentation toassure that it can be used without concern of cross contamina-tion. Recovered solvents must meet predetermined specifica-tions. Where recovered or recycled solvents are used in themanufacturing process, their use must be documented.

When customer returns are received, the material mustbe placed in quarantine to separate them from approvedmaterials. The reason for the return, as well as investigationinto the cause for this reason, must be conducted and docu-mented. The material must be evaluated to determine if thequality of material is affected and=or if the material can bereturned to stock. Additional testing may be necessary inorder to make that determination. Any testing must bedocumented. If reprocessing or rework is necessary, it mustbe performed in accordance with the requirements detailedabove.

Where materials have been exposed to extraordinaryconditions such as extreme temperatures, smoke from a fire,radiation from natural disaster, or other similar incidents,that material should be destroyed. Testing should be designedto be appropriate for the use of the material.

a. Compliance Requirements

All reprocessing and reworking must come to the atten-tion of the quality unit for final review and approval. Contin-uous reprocessing to bring a batch into conformance shouldnot be allowed, as it indicates that there is something unu-sual and unknown about the process and/or the quality ofthe product. The need for additional or new tests must be

Quality Assurance and Control 271

Page 287: Active Pharmaceutical Ingredients

decided before a reprocessed batch can be released. The qualityprofiles of reprocessed or reworked batches should be comparedto normal, first-time-right batches. This should include purityand impurity profiles, as well as physical profiles. In general,all rework processes require prior regulatory approval.

For each customer return, document the investigationinto the reason and cause for the return. Identify correctiveactions where appropriate. The quality responsible personshould make the decision to return material to stock, furtherprocess it, or discard it. While this is not spelled out in theregulations, it should be clear that only the quality unit hasthe authority to return materials to production, as this servesas a release function.

15. ICH Q7A Section 15: ‘‘Complaints andRecalls’’

Customer complaints represent a unique opportunity forquality improvement. Complaints can come into a companyin a variety of ways: person-to-person, via a telephone call,through e-mail, or regular mail. All quality related com-plaints received by any employee of the firm must be chan-neled to the quality department, investigated, and thisinvestigation must be documented. The record must mini-mally contain the name and the address of the person initiat-ing the complaint, the date the complaint was received, and adescription of the complaint. The investigation must includethe final decision regarding the material and a copy ofthe response sent to the complainant. Complaints shouldbe periodically reviewed for trends suggesting areas ofimprovement.

If the complaint is of a serious nature, which might jus-tify concern of the material on the market, local regulatoryagencies must be contacted within time frames stipulated inthe local requirements. In rare occurrences, recall of mar-keted products may be necessary. Each firm must have inplace procedures that define how a recall is to be conducted.The person responsible for the recall must be identified inthe procedure.

272 VanderZwan

Page 288: Active Pharmaceutical Ingredients

a. Compliance Requirements

All complaints must be investigated and documented bythe quality department. The recall procedure is one procedurethat a manufacturer does not want to gain experience inimplementing. However, when a firm finds itself in a recallsituation, decisions must be made quickly and a well-definedprocedure can facilitate the communication and decision-making processes. The procedure should identify the sourcesand types of information necessary for the decision, whichfunctions are to be present in an advisory capacity, and theultimate decision maker whether to proceed with a recall.

16. ICH Q7A Section 16: ‘‘ContractManufacturers (Including Laboratories)’’

When another firm manufactures or tests products or materi-als, the responsibilities identified in this chapter must bedefined as falling under the contract giver or the contractacceptor. A formal document typically captures the assign-ment of these responsibilities and is termed the quality agree-ment. Each contract manufacturing or testing situation isunique and will require a quality agreement specifically tai-lored for that situation.

a. Compliance Requirements

The contract giver and the contract acceptor each usuallyhave their own template for their quality agreements. Thuswhen a contract-manufacturing situation is entered into,the assignment of responsibilities and capturing these into aquality agreement requires negotiation not only for theresponsibilities but also the formatting of the agreementitself. There is no right way except to insure that there isclarity from both parties as to the responsibilities.

IV. THE QUALITY CONTROL AND QUALITYASSURANCE DEPARTMENT

The cGMP regulations define the responsibilities of the qual-ity control and quality assurance department throughout all

Quality Assurance and Control 273

Page 289: Active Pharmaceutical Ingredients

phases of manufacturing. Part IV of this chapter on qualityreviews the specific laboratory controls, and all the QC=QA-related responsibilities throughout the regulations.

A. Laboratory Controls, Taken from the FDA’sRegulations for cGMP

The first subsection covers general controls. As expected, allactivity associated with the testing of materials must be scien-tifically thought out. Sampling must be based on statisticalgrounds, and all procedures should be documented. Thisincludes all activities of the laboratory in its efforts to evaluateall materials, from rawmaterials to containers, intermediates,in-process controls, and so on, through to the stability testingof the final APIs.

Testing for the release of final products should be per-formed on each lot produced. Sampling plans, based on statis-tical grounds, should also include supportive data illustratingthat the batch is homogeneous and the process will alwaysyield a uniform grade of material. The test methods must bevalidated, which means their accuracy, sensitivity, and line-arity over a variety of concentrations of material, specificity,and reproducibility have been established. Such criteria applyto all test methods used throughout the manufacturing pro-cess, not only to the API.

Stability testing is also required to demonstrate that thematerial will hold its quality over the labeled storage condi-tions and time. When establishing the storage conditions forthe first time for an API, studies should include the extremesof conditions likely to be seen. The testing protocol shouldinclude all and only those attributes that may be affected bythe storage conditions. Consideration should also be given tothe stability of the product during its planned method of ship-ment. The regulations offer an adequate amount of flexibilityto the storage conditions for the stability study samples. Therequirement logically states that the sample container affordsthe same level of protection, as does the bulk container. Theresults from these studies are used to determine either anexpiry date or a re-evaluation date. Re-evaluation dates are

274 VanderZwan

Page 290: Active Pharmaceutical Ingredients

preferred since APIs exceeding their expiry dates are to bediscarded, while those with re-evaluation dates may bereturned to stock following a satisfactory re-evaluation.

As APIs are tested for purity, impurity testing on a lot-by-lot basis is also required. The expected impurities shouldbe determined for normal production batches, and new impu-rities should be hunted down when evaluating a processchange.

A sample from each lot of API and key intermediateshould be taken and stored for annual visual examination,as well as to cover any future investigative purposes (for exam-ple, to evaluate a customer complaint on the lot).

Finally, if animals are used for testing purposes (althoughthis is very rare today), they require the same degree of controland suitability for testing as analytical equipment, reagents,and other aspects of the manufacturing process.

1. Compliance Requirements

Laboratory operations must be documented, and the QC lea-der must approve any changes. Retesting or resampling is aserious matter that can only be conducted following specificconditions and must be carried out under predescribed writ-ten procedures. See FDA’s Guidance for industry ‘‘investigat-ing out of specification test results for pharmaceuticalproduction,’’ issued on their website (fda.gov) September 4,1998. An effective calibration program for all equipmentand reagents is required. The source of reference standards,their storage, and use should be clearly defined. All differentphysical forms of an API must be included in the routinestability study program. Manufacturing change control sys-tems should evaluate all likely attributes that may beaffected, as well as additional attributes that are not part ofa typical release protocol (for example polymorphism or newimpurities). Initial lots from a process change should be addedto the stability study program. An effective program tovisually examine each lot of each key intermediate and APImust be established, the results recorded, and any cause forinvestigation taken are also documented.

Quality Assurance and Control 275

Page 291: Active Pharmaceutical Ingredients

B. The Quality Unit Responsibilities

1. The Reporting Relationship and GeneralResponsibilities

Throughout the cGMPs, reference is made to ‘‘the qualityunit.’’ That is the way the FDA and other agencies addressthe areas of the organization responsible for the quality con-trol, quality assurance, and other QM activities. While theregulations do not mandate to which area or departmentthe quality unit should report, all regulators make it clearthat they must have a reporting relationship that allows,and even encourages, independence of judgment. The peopleassigned the responsibility to judge the quality of a product,material or process cannot be expected to do a good job forthe company if there is a conflict of interest between themand their direct supervisors. Regulators and company seniormanagement agree on this point because we are dealing withthe manufacture of products used to treat human illness anddisease. There is no margin for error.

The overall responsibility of the quality unit is to helpthe organization develop and implement a solid system of pro-cedures and controls to ensure that each batch of API will rou-tinely meet its predefined quality attributes. To execute thatresponsibility, the quality unit inherits a broad range ofauthority: to review and approve all procedures, all systems,all changes, all in-process and final product specificationsand test methods, all manufacturing procedures, investiga-tions, and so on. The full scope of authority can onlybe appreciated by a thorough reading of the FDA’s cGMPdocument. Suffice it to say that all activity related to the man-ufacture of APIs require the approval of the quality unit.

Clearly, top scientists, technicians, and managers areneeded in the quality unit in order to facilitate a smooth-running manufacturing organization.

2. The Quality Control Department

The general laboratories of the quality unit are often referredto as the QC department. This is the area of the quality unit

276 VanderZwan

Page 292: Active Pharmaceutical Ingredients

responsible for carrying out the tests on the purchased mate-rials, in-process samples, intermediates, final APIs, and stabi-lity studies. In some cases, parts of this testing can bedelegated to other departments, such as in-process controltests to the production area. However, the final release deci-sions are still the responsibility and authority of the qualityunit. The quality laboratories are considered part of the man-ufacturing plant; the QC function is part of the manufactur-ing process and comes under the same regulations asproduction areas. They therefore require that systems bedescribed in writing, that cGMP training occurs on an ade-quate basis, and so on. The test procedures used in thisdepartment must be validated. The equipment used must bequalified to demonstrate it functions as designed, and it mustbe maintained and calibrated on a sufficiently regular basis toassure that it is always working properly. Any problems thatoccur must be investigated and corrected.

3. The Quality Assurance Department

This part of the quality unit is responsible for the review andapproval of the cGMP written procedures and systems usedthroughout the site for the manufacturing, control, and therelease of API’s. The QA department typically has responsibil-ity to write quality policy and standards, and to prepare SOPsfor the quality control and quality assurance department tofollow. The quality unit typically has the responsibility toaudit the manufacturing and QC functions to assure thatthey are following their procedures correctly and they arecompliant with other aspects of the regulations, such as per-forming and documenting investigations where necessaryand implementing corrective measures where appropriate.

Another major responsibility of this department ischange control management. To effectively evaluate thepotential impact of a process change, it is important to con-template how the predefined quality attributes might beaffected—as well as other chemical=physical attributes notnormally tested or evaluated. This requires people who havea firm understanding of the chemistry of the process and an

Quality Assurance and Control 277

Page 293: Active Pharmaceutical Ingredients

appreciation of how a change in the API may affect the drugproduct process throughout the supply chain.

The significance of the responsibilities of the quality unitand the scope of its influence throughout the manufacturingprocess, only briefly highlighted in this section, requires thatit be staffed with very well-educated, -experienced and -skilled people who are good thinkers, communicators, andconfident decision makers.

Other duties of this function usually include the hand-ling and investigation of customer complaints, cGMP train-ing, throughout the manufacturing site and review andapproval of major projects such as validation reports or capi-tal investments to assure or improve cGMP compliance.

4. Analytical Technical Service

An analytical technical services department should also existwithin the quality unit. The functions of this departmentinclude helping manufacturing in troubleshooting to deter-mine ‘‘root causes’’ for quality problems, improving currenttest methods to make them more efficient or more userfriendly, evaluating new technology, evaluating inquiriesfrom official offices such as pharmacopoeia, performing analy-tical investigations to evaluate complaints against quality,and to keeping all current test procedures up to date.

5. Management of Quality

The head of the quality function has the overall responsibilityfor the quality systems at the site or across the company.He or she is not the single responsible person to produce aquality product; that responsibility belongs to the head ofmanufacturing. This distinction may not always be clearlyunderstood. To delineate the division of responsibility moreeffectively, the head of quality is responsible for ensuring thatthe requirements are effectively communicated and under-stood by plant management and an effective compliance andquality system is developed and implemented to achieve thoserequirements. It is the responsibility of the head of manufac-turing to ensure that the system is properly supported,

278 VanderZwan

Page 294: Active Pharmaceutical Ingredients

financed, and strictly adhered to. The head of quality, on theother hand, has the sole responsibility to release the productfrom the plant for further processing, or for sale to a drug pro-duct manufacturer. Through the authority to audit, the qual-ity head learns if the systems are being followed; if not, it ishis or her responsibility to bring that matter to the attentionof the head of manufacturing. If corrective measures are notforthcoming as soon as required based on the severity of theobservation, the quality leader is responsible for adequatelycommunicating the matter to a higher level in the organiza-tion, regardless of ‘‘lines of reporting’’ as described in organi-zation charts.

A further role of the quality leader is to encourage supportand enthusiasm for quality and quality improvement through-out the entire organization. This is best conducted if the qualityleader solicits the support and confidence of themanufacturinghead, as well as all other technical management at the site andamong senior leaderships throughout the organization.

Finally, the quality leader must ensure that his or herstaff have access to current information, practices by othercompanies, and enforcement efforts by the regulatory autho-rities. This is necessary in order to ensure that the manufac-turing system continues to keep pace with the ‘‘current’’component of the cGMPs.

Quality Assurance and Control 279

Page 295: Active Pharmaceutical Ingredients

Appendix

ATypicalAttributesEvaluatedto

Con

trol

andAssure

APIQuality

Attribute

How

determined

Specification

example

Ration

ale

Description

Visualex

amination

Agrees

withstandard

ortypicalproduct

description

Atraditionalorganolep

tic‘‘test’’

withverylimited

valuedueto

itssu

bjectivity,nev

er-the-less,it

remainsin

use,simply

toprovidetheassurance

thatthe

batchappea

rssimilarto

that

expected

Iden

tity

IR,NIR

,FTIR

,NMR,

UV,MS,etc.

Agrees

withstandard

Theseelectrom

echanical =

spectroscopic

analyseshelp

verifythatthecorrectch

emical

bon

dsandarrangem

entof

functionalgroupsare

present

andelucidate

thestru

cture

ofthemolecule

Watercontent

Karl

Fisch

erVariable,reportedas%

by

weight,

comparesto

normal

ranges

TheKarl

Fisch

ertest

determines

how

much

wateris

presentdue

toinefficien

tdrying,the

hygroscop

icnature

ofthe

molecule,and=or

isch

emically

bou

ndasahydrate,butdoe

snot

distinguishbetwee

nthesetypes

Residual

solven

tsGC

Typicallyreportedas‘‘n

otmore

thanX

%’’and,ifthesolvates

are

controlled=sp

ecified

inan

Determines

how

much

residual,or

volatile,process

solven

tsadhere

tothemolecule

from

inefficien

t

280 VanderZwan

Page 296: Active Pharmaceutical Ingredients

officialpharm

acopoe

ia,their

limitswou

ldbesp

ecified

dryingand=or

asabou

nd

solvate,butdoe

snot

distinguish

betwee

nthetw

otypes

Crystallinity=

morpholog

yX-raydiffraction

Con

form

sto

diffractog

ram

ofdesired

polymorph

Theou

tput,

called

a‘‘diffractog

ram,’’

provides

veryprecise

mechanicalinform

ation

abou

tthe

shapeof

thecrystal,

reportingthree

dim

ension

sof

the

crystal’saxis

andthe

arrangem

ent=pack

ing

ofthemolecules

within

thecrystal

Meltingrange

Differentialscanning

calorimetry

Thermog

ram

andmelting

curveconform

todesired

polymorph

Theou

tput,

called

a‘‘thermog

ram,’’

provides

an

indication

ofhea

tabsorp

tion

ofthemolecule

duringthemelting

phase;acomparisonto

areference

standard

ofthe

product

provides

anindication

ofthesa

mple’s

purity

Particle

size

distribution

Microscop

y,laser

obscuration

,laserlight

scattering,laser

diffraction,etc.

Profile

ofdistribution

matches

typicalresu

lts

Theou

tputis

adistribution

ofparticle

sizes,

show

ingthe

percentpresentatev

erincrea

singsizes,

andarange

andmea

n,reportedin

microns

(Con

tinued

)

Quality Assurance and Control 281

Page 297: Active Pharmaceutical Ingredients

Appendix

ATypicalAttributesEvaluatedto

Con

trol

andAssure

APIQuality

(Con

tinued

)

Attribute

How

determined

Specification

example

Ration

ale

Hea

vymetal

content

USPmethod

sTypicallyreportedasless

than

Xppm

Indicatesthepartsper

million

of(poten

tiallytoxic)hea

vymetal

contaminants=im

purities

inthe

dru

gsu

bstance

Palladium

(platinum),

etc.,

content

AA,IC

P-O

ES,IC

P-M

STypicallyreportedasless

thanX

ppm

Indicatesthepartsper

million

ofsp

ecificallypalladium

orplatinum

(oranysp

ecificmetal

when

usingtheappropriate

detector)

inthedru

gsu

bstance.

Asp

ecificmetaltest

isconducted

when

ever

asp

ecific

metalis

usedasacatalyst

duringthesynthesis.

Chiralpurity

HPLCor

opticalrotation

%en

antiom

eric

excess;or

deg

rees

ofsp

ecificrotation

,comparedto

chirallypure

reference

standard

Provides

anindication

ofthe

enantiom

eric

purity

ofach

iral

material,this

test

isconducted

when

thedru

gis

designed

toex

istasasingle

enantiom

er,or

when

anen

antiom

ericallypure

chiralraw

materialwasusedin

thesynthesis,andthech

irality

ordeg

reeof

enantiom

eric

purity

isdeliberately

destroy

edto

yield

aracemic

mixture.In

this

case,

thesp

ecification

wou

ldbe,

for

282 VanderZwan

Page 298: Active Pharmaceutical Ingredients

example,less

than2%

enantiom

eric

excess

Impurity

HPLC

Lim

itsbasedon

process

averages,sa

fety

and=or

regulatory

expectation

s;gen

erallyreportedastotalis

less

thanX%,noknow

nim

purity

greaterthan0.5%;no

unknow

nim

purity

greater

than0.1%

(orlower–

sometim

esmoredetailed

)

Theim

purity

profile

isvery

importantbothasanindicator

ofthepurity

aswellasash

owof

consisten

cyandreliabilityof

the

manufacturingandpurification

processes

Assay

HPLC

Not

less

than98%,not

more

than102%,Com

paredto

reference

standard

ofknow

npurity=poten

cy(w

eight=weight

basis,

ondried

samples)

Proof

thattheprocess

yieldsthe

expectedpurity

Microbial

purity

Viable

aerob

iccounts,

yea

stsandmolds,

specificindicator

organisms

See

USPor

EP

Microbialtestsrevea

lthe

bioburd

enassociatedwiththe

dru

gsu

bstance.

Biomaterials

gen

erallycome

from

process

waterusedin

thefinalstep

.Thetest

isnot

necessa

ryifthefinalstep

innon

-aqueo

us,

andthedrying

andsu

bsequen

thandlingare

controlledsu

chthat

bio-exposure

islimited

ornot

possible

Quality Assurance and Control 283

Page 299: Active Pharmaceutical Ingredients
Page 300: Active Pharmaceutical Ingredients

8

Plant OperationsSTANLEY H. NUSIM

S. H. Nusim Associates Inc., Aventura, Florida, U.S.A.

I. Plant Organization . . . . . . . . . . . . . . . . . . . . . . 285II. Batch vs. Continuous . . . . . . . . . . . . . . . . . . . . 286III. Dedicated vs. Shared Manufacturing Facilities . . . . . . 288IV. Shift Operations . . . . . . . . . . . . . . . . . . . . . . . 288V. Sterile Operations . . . . . . . . . . . . . . . . . . . . . . 291VI. Clean Room . . . . . . . . . . . . . . . . . . . . . . . . . . 294VII. Cost Control . . . . . . . . . . . . . . . . . . . . . . . . . 296VIII. Fixed Overhead Absorption . . . . . . . . . . . . . . . . . 299IX. Safety . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300X. Environmental . . . . . . . . . . . . . . . . . . . . . . . . 303

I. PLANT ORGANIZATION

The first step is the planning of the organization of the plant.I will, in this discussion, make certain assumptions about thecompany. Many of the necessary activities required for apharmachemical business operation, other than the pureproduction operations themselves, should usually be theresponsibility of a separate organization within the company.

There are usually four specific organizational areasrequired in any plant organization. These are the manufac-turing operations themselves, plant maintenance, materialsmanagement and quality control and assurance. The lastthree activities are the subject of individual chapters in thisbook and I refer the reader to those. I will concentrate onthe manufacturing operations and restrict my comments to

285

Page 301: Active Pharmaceutical Ingredients

broad generalities on these allowing the full chapters to coverthe appropriate details.

Other key activities, also the subject of separate chaptersin this volume, relate to process development, plant design,and regulatory activities that are all critical to the initiationand approval of any plant. However, these activities in anylarge organization are separate functions that interact withthe plant. Process development as well as regulatory opera-tions is traditionally part of a corporate research division,while plant design would generally be part of a corporateengineering function.

The interaction of the plant with the research and devel-opment area will be most significant during the design andstartup phase of the plant and each of its products. The regu-latory activities that would be required for the preparationand filing of the required documentation for the FDA, in theUnited States, or the Health Ministry of the country wheresales are planned will be the subjects of discussion in theappropriate chapter.

II. BATCH VS. CONTINUOUS

The evolution of the industry has seen an interesting shift. Theearly days of pharmachemical manufacturing, particularly inEurope, were focused on the input of ‘‘industrial chemists’’who developed the complex multistep processes often requiredto achieve the unique structure of the product. This, in combi-nation with the relatively small commercial requirements, ledto relatively small-scale batch operationswith batch equipmentin the 100–500-gal size. Generally, the equipment was a rela-tively small scale up from the laboratory and mirrored thelaboratory operation. This was more than adequate to meetthe then small market needs. In addition, new product produc-tion requiringdifferent equipmentneedswould bemet by build-ing another small factory within the plant and resulted in aplant maintaining many small dedicated operating factories.

As the business of pharmaceuticals grew, particularly inthe 1940s and 1950s, the kilogram requirements for the

286 Nusim

Page 302: Active Pharmaceutical Ingredients

specific pharmachemical leapt. This drove the need for largervessel volume and more complex batch systems and this drovethe capital cost up at a time when equipment costs werealready rising.

Furthermore, during these decades other impacts beganto weigh on the capital and operating costs of each and everyfactory in a plant. These were the new issues of environmen-tal impact and control and the increased safety and personnelexposure control mandated for each operating unit. All ofthese factors increased the cost to operate each separate unit.This gave impetus to allow the growth of the impact of the che-mical engineer, rather than the chemist, on the transfer of thesynthetic process from the laboratory to the commercial scale.In these years, particularly in the United States, the applica-tion of more traditional large-scale continuous manufacturingtechniques already in use in the high-volume petrochemicaland heavy organic chemical industry was introduced. Thisbecame possible only because of much larger requirementsfor the pharmachemical from thousands to millions of kilo-grams per annum. In addition, continuous processing wasoften superior to batch processing because of the increasedprocess efficiencies that were possible and, very significantly,the ability to limit time and temperature conditions to themore temperature-sensitive molecules now often beingdemanded for the newer products.

Surprisingly, the increased material requirements, dri-ven by explosive product volume growth, have seen a compen-sating effect in recent years. Although the growth of marketvolume continues to increase requirements for product, thenew research techniques developed in the 1980s and 1990shas resulted in much more potent products greatly reducingthe quantities of pharmachemical needed for the market. Asan example, the first antihypertensive drug,methyldopa, initi-ally marketed in the 1960s, had a 250–500 mg dose regimen.Hence, for each billion tablets required for the market about400,000 kg of drug was needed. Today, the most popular med-ications to control blood pressure are ACE inhibitors, whichrequire only a 5, 10, or 20 mg dose regimen. Thus, for 1 billion10 mg tablets, only about 10,000 kg of bulk drug is needed.

Plant Operations 287

Page 303: Active Pharmaceutical Ingredients

The net result of this is the refocus on batch operationsfor most APIs.

III. DEDICATED VS. SHAREDMANUFACTURING FACILITIES

A number of factors have led to the shift from dedicated to thesharing of manufacturing facilities for APIs. The first is,again, the increased potency of APIs and hence the reducedquantities required satisfying the market requirements.

The second point is the significant capital cost require-ments for a facility. This cost increase starts with normal infla-tion trends over the years and has added to it a number of otherfactors. The first is the increased sophistication of the chemis-try that accompanies the increasingly complex chemical moi-eties being synthesized on a commercial scale. Today, whathad been strictly laboratory procedures and unit operationssuch as chromatography has become a routine plant operation.

In addition to these, more nontraditional operations on acommercial scale, are the sharply increased requirements inmost countries focusing on cGMP. One then adds as well tothe operational issues that of waste management, environ-mental concerns, and safety and employee exposure.

The final point is the normal economics of larger-scaleproduction. If one continued to utilize dedicated facilities foreach product and was even willing to spend the capitalneeded, they would be faced with the higher unit operatingcosts associated with smaller batch sizes required to operatefor each API that resulted from the reduced requirements ofAPIs. Utilizing the same facility for multiple products dictatesthe need for larger batches and the resultant economy ofscale.

IV. SHIFT OPERATIONS

The chemical manufacturing industry has long seen the needfor round-the-clock multishift operation.

288 Nusim

Page 304: Active Pharmaceutical Ingredients

Initially, batch chemical processing saw a need forextending shift operations beyond one shift operation onlywhen the process needs dictated.

However, as the chemical industry and later the phar-machemical industry extended to continuous operations24 hr round the clock, multiple shifts became common oftenextending to 7 days.

Later, as the return to batch processing occurred in morerecent years, the idea of multishift operation was maintainedfrom an efficiency point of view in order to more effectivelyuse the invested capital in the plant.

There are a number of approaches to scheduling shiftwork for a 5-day around the clock operation. The simplest,administratively, is to have people assigned to the same shiftroutinely. This may be efficient to supervise but it forces afixed day, afternoon, and night ‘‘crew’’. Keeping people on anight-only schedule is difficult to sustain as turnover wouldbe high and would be generally more difficult to maintainfrom the employee perspective.

This leads to the more common and generally more accep-table three-shift rotating arrangement whereby each weekthe shift crews are rotated, first from day to afternoon andthen, the following week, from afternoon to night shift. Thisis a more reasonable schedule that has been used routinelythroughout plant operations.

The more administratively challenging schedule is tooperate on a 24-hr 7-day schedule. Clearly, this is the mostefficient of all schedules because it uses all 168 hr in theweek without stoppage. The elimination of stoppage itselfis an added efficiency as there is always some lost timewithin the scheduled work program whenever a shutdownoccurs. As an example, even in a 5-day 24-hr operation, asthe end of the fifth (and final) workday there must be somelost time associated with the facility shutdown for theweekend, as well as time lost on the first day to restartthe operation.

The normal ‘‘stagger shift’’ operation I have seen andused involves four ‘‘shift crews’’ who work 8 hr a shift begin-ning at 8AM (day), 4PM (afternoon), and midnight (night).

Plant Operations 289

Page 305: Active Pharmaceutical Ingredients

They work a full 7 days in succession on each shift with daysoff between shifts varying from 1 to 4 days. This system,outlined in Table 1 operates on a repeating 28-day cycle.Thus, each employee works 21 days out of 28, working 1 fullovertime day per cycle.

This operational system, or variations of it, is commonlyused. Its advantage is that it allows for overlapping coveragefor sickness, training sessions, and vacation. Thus, if a personis unable to fill his=her shift for any reason the surroundingpeople each work an additional 4 hr. This permits operationwithout outside (less experienced) people stepping into anunfamiliar operation. This is important in the manufactureof a regulated drug product, where familiarity and under-standing of the operation is critical.

More recently, somewhat driven by the issue of energy con-servation that emerged during the fuel crises of the 1970s is a12-hr shift cycle, one shift starting at 8AM and the second at8PM.This requires four crews thatwork 4days on and 4days off.

The example is given in Table 2.This shift arrangement requires much more respon-

sibility as overlapping cannot fill absences, as it would require18-hr shifts. It requires more flexibility to cover vacancies.

Table 1 Seven-Day Stagger Shift

DayCrew 1 2 3 4 5 6 7 8 9 10 11 12 13 14

I D D D D D D D O N N N N N NII A A A A A O O D D D D D D DIII N O O O O A A A A A A A O OIV O N N N N N N N O O O O A A

DayCrew 15 16 17 18 19 20 21 22 23 24 25 26 27 28

I N O O O O A A A A A A A O OII O N N N N N N N O O O O A AIII D D D D D D D O N N N N N NIV A A A A A O O D D D D D D D

D, Day shift; A, afternoon shift; N, night shift; O, off.

290 Nusim

Page 306: Active Pharmaceutical Ingredients

However, it is very desirable from the worker’s perspective asit provides long breaks every week.

V. STERILE OPERATIONS

In the preface to this book, it is pointed out that fermentationoperations will not be dealt with in this volume. This wouldleave the great bulk of sterile operations out of this text; how-ever, there are some sterile operations that must be consid-ered. However, I would not represent this volume to be anauthority on this aspect of pharmachemical processing. Morespecific references should be pursued by the reader.

The primary focus here will be in synthesized pharma-chemical that would go into two types of pharmaceuticalproducts; parental drugs, such as antibiotics or ophthalmicwhere regulatory authorities require sterile products fortopical treatments involving the eye.

In the discussion of sterile operations, one must firstrecognize that there are two distinctly different reasons forsterile operations. The first is product contamination, particu-larly if the pharmachemical is used directly as an injectable orintravenous material. Here it is necessary to assure that thereare no living organisms present that could pose a threat to thepatient. The second is completely different; if, as in fermenta-tion or other biological processing, one is carrying out a biolo-gical process one must be certain that no foreign livingorganism are present during that operation. The efficiency aswell as the viability of the organism itself could be affectedby the presence of an unplanned biological component. In this

Table 2 Four-Day Stagger Shift

DayCrew 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16

I D D D D O O O O N N N N O O O OII N N N N O O O O D D D D O O O OIII O O O O D D D D O O O O N N N NIV O O O O N N N N O O O O D D D D

D, Day shift; N, Night shift; O, Off.

Plant Operations 291

Page 307: Active Pharmaceutical Ingredients

volume, we are not addressing the biological processing opera-tions; thus, our focus will be on those synthetic chemicals (asan example antibiotics that will be injected into people).

The approach to achieving sterility in a finished productcan be achieved in two different ways. The first is asepticprocessing and the second is ‘‘terminal’’ sterilization. Asepticprocessing will be discussed here, as terminal sterilization isfavored as amore certainmethod as it eliminates any potentialcontamination during processing.

A. Terminal Sterilization

The processes used for terminal sterilization are heat, tempera-tures in excess of 140�C, gamma ray radiation, perchloric acidfumes, and ethylene oxide. These techniques are commonlyused for sterilizing metals and plastic tools, medical devices,and instruments. This approach is clearly preferred as it pro-vides a final step that assures sterilization of the product andonly demands limited controls prior to that final step. However,unfortunately synthetic organic chemicals, particularly syn-thetic antibiotics are generally not able to accept these severeconditions without adverse effect on the product. This leads tothe need to pursue aseptic processing.

B. Aseptic Processing

The concepts of aseptic processing for pharmachemicalprocessing to assure the sterility of the product is no differentthan classical parental drug production. A sterile facility has anumber of fundamental characteristics; the first is completeseparation and isolation from all other operations. This dic-tates that people, materials, and even the ventilation be inde-pendent of all other activities at the site. First, the HVACsystemmust be totally independent of the main system, it pro-viding only filtered air, generally through HEPA filters thatremove essentially all particulate matter larger than 0.21 mm.

Levels of cleanliness have been established as ‘‘classes’’,an industry convention. The ‘‘class number’’ measures themaximum number of particles per cubic meter that may bepresent. It does not represent the number of organisms.

292 Nusim

Page 308: Active Pharmaceutical Ingredients

Particles are more easily and uniformly measurable thanorganisms and it is assumed that as the number of particlesgrow the opportunity for some of these either being organismsor having organisms on them increases.

The general standard for a sterile environment in thepharmaceutical industry is Class 100. The general standardfor pharmaceutical processing is Class 100,000, while oftenthe preparation area servicing a sterile area would normallybe Class 10,000. The purpose of this somewhat tighter stan-dard for a sterile preparation area is to reduce the burdenon the Class 100 sterile systems that it services. It shouldbe noted that the electronics industry that deals with micro-chips is even more concerned with particulate matter(whether or not they are living organisms) and has an evenmore stringent requirement, Class 10.

Access is restricted to materials that have previouslybeen sterilized by classical sterilization. People gain accessonly following a complete gowning including feet and headcovering. The facility itself must include smooth walls andfloors that are easily washable and equipment that must becapable of disassembly for cleaning. It is possible to avoidequipment disassembly by installing sterilizing systems inplace of the larger equipment. Each of these sterilize-in-place(SIP) systems must be validated to assure its effectiveness.

There are procedures that must be followed in extremedetail without exceptions in order to assure continuing sterilityand limiting people access (the greatest source of organismcontamination are people). A facility if used continuously in asterile condition requires a periodic resterilization. Historyand validation would dictate the frequency of the need to rest-erilize. Obviously, if sterility is broken, e.g., by an equipmentreplacement or even a breakdown of the HVAC system, rester-ilization would be required before processing could resume.

As can be deduced from the above, a sterile operation ismuch more costly both in operating expense as well as origi-nal capital outlay than a traditional nonsterile pharmachem-ical facility. Hence, one can quickly see the merit of terminalsterilization that eliminates all of these added burdens foraseptic operation. However, if terminal sterilization is not

Plant Operations 293

Page 309: Active Pharmaceutical Ingredients

feasible, then one should work one’s way back from the pureproduct until a point is reached in the process where defini-tive sterility can be achieved. This will limit the added opera-tional burden and cost required for aseptic processing.

Often this is possible in the very last process step. Mostoften once the desired pharmachemical is formed, it is thenput through a recrystallization step to achieve the high puritylevel usually demanded. The key is for the crude material tobe put into solution and either simply crystallized or carbontreated and then crystallized. A natural opportunity is whenthe material is dissolved in the final step. The solution canthen be passed through a submicron sterile filter into an asep-tic environment where new contamination cannot occur. Thislimits the size and scope of the sterile facility and its asso-ciated premium costs.

If such a solution step were not in the process, then onewould have to follow the procedure described above to locatethe latest point in the process where a sterile filtration canbe achieved. Considering that all processing subsequent to thatsterile filtration stepwould have to be carried out under asepticconditions, it might be more cost effective to add a sterile filtra-tion step at the end of process, then operate a number of stepsprior to the end of the process. Clearly, a cost study should becarried out to determine the optimum course of action.

VI. CLEAN ROOM

Today, in the physical plant probably the most significant dif-ference between a traditional chemical facility and a pharma-chemical facility is the ‘‘clean room’’ in the pharmachemicalplant.

This concept, introduced in the 1970s is derived from thelandmark U.S. FDA regulation that established ‘‘current goodmanufacturing practices.’’ This legislation did not in itselfdefine the need for a clean room, but it essentially requiredit in order to be in compliance.

Fundamentally, the legislation did two things: redefinedwhat is a contaminated product and shifted ‘‘quality’’ from‘‘quality control’’ to ‘‘quality assurance.’’

294 Nusim

Page 310: Active Pharmaceutical Ingredients

Up until the introduction of cGMPs, a ‘‘contaminated’’product was defined by quality control testing showing that:either the material did not meet the specifications or thatthere was seen in the testing the presence of a contaminant.This, of necessity, presumes that contamination is uniformlydistributed in the product and that the samples tested arerepresentative of the batch.

This ‘‘uniform’’ contamination concept worked in thecases that contamination occurred in the processing itselfand was, therefore distributed throughout the product. How-ever, it did not deal with extraneous contamination that couldcome from the room environment or the equipment thatwould not be chemical contamination as would easily bepicked up by testing. This type of contamination would berandom in nature and not subject to normally applied sam-pling techniques.

Thus, the new legislation defined strict ‘‘good manufac-turing practices,’’ which provided detailed rules governingthe controls that must be in place in the facility, the equip-ment, the people, the documentation, and the process. Itessentially said that if a factory was found to be out of compli-ance to the GMP requirements, it could not assure that anybatch of product was free of contamination; hence, all batchesare declared contaminated, regardless of the testing resultsfor any batch.

In terms of facilities, the key issue was the commonfactory environment. It was the practice of isolating the finalAPI in essentially the same environment as all prior inter-mediates. This potentially exposed the final API product tothe same factory environment, where extraneous contamina-tion ranging from paint chips from equipment to rust fromoverhead piping and unfiltered solids from earlier processsteps could carry forward into the final product.

The most practical way to meet this new requirementwas to provide a separate ‘‘clean room’’ within the factorywhere the environment would be subject to higher levels ofcontrol than in the normal operating area.

I had the opportunity of overseeing the establishment ofthe first ‘‘clean room’’ at Merck’s multiproduct plant in

Plant Operations 295

Page 311: Active Pharmaceutical Ingredients

Rahway, New Jersey, in the late 1970s. The facility wastailored after a sterile room concept where access was limited,clean uniforms required with the appropriate hair covers. Ithad controlled and separated air-handling systems, appropri-ate wall and floor finishes, no protruding piping or fixtureswhere dust or dirt could collect, and thorough cleaningprocedures.

It was designed to be the finishing room where the finalisolation of the API took place and any subsequent finishingsteps such as milling, final blending (required for allproducts), and packaging. Even within this controlled envir-onment, covered containers were used whenever transfer ofmaterial was necessary.

The size of the area and the extent of processing thatwould be carried out in the clean room would be dictated bythe process. Logically, the best approach would be passing asolution of the product through a fine filter and taking itthrough a separating wall into the next vessel that is locatedin the clean room. Normally, most products have a recrystal-lization step that would fit the need. In the unusual eventthat a recrystallization is not part of the process, one wouldgo back into the process to a point where a solution of an inter-mediate exists. It would then require all subsequent steps tobe carried out in the clean room. This could cause a significantincrease in cost to erect an expanded clean room. One shouldconsider whether adding a standard recrystallization to theprocess could be justified as capital avoidance.

Another significant decision is whether to build a dedi-cated clean room for each product or minimize capital by hav-ing a single clean room for all products at the site. This wouldbe generally cost effective for multiproduct factories, particu-larly if the products have relatively small volumes. Again, theoverall economics would govern.

VII. COST CONTROL

The costs of any business operation must be monitoredand controlled. A pharmachemical factory is no exception.

296 Nusim

Page 312: Active Pharmaceutical Ingredients

The usual costs incurred can be broken into a variety ofclassifications that I will discuss later.

It is clear that if the plant operates to produce a singleproduct then regardless of the cost system used, the actualall inclusive manufacturing costs incurred will be the basisfor the product gross margin (PGM), which is the sales rev-enues minus the actual manufacturing costs. This is straight-forward and does not pose special problems.

In a plant, however, where more than one product ismade, one must develop a control to allocate the shared coststo the various products. This is not as simple as it sounds andcan influence the perception of the profitability of a product.

The first consideration is to determine the fixed andvariable costs involved with every product manufactured.Some items are obvious, others are not. A variable cost isdirectly proportional to the quantity of a product manufac-tured such as raw materials and auxiliary chemicals used inthe synthesis. For each kilogram of product, a specific amountof each raw material is required. If production of that productceased, there would be no expense for that raw material.Similarly, the labor used to run the batch is also variable;the labor used is directly proportional to the number ofbatches run. On the other hand, an essentially fixed cost isindependent of the quantity of a product made. This wouldinclude supervision at the factory level and clean out chemi-cals used for turn around of the equipment. However, thereare many classes of costs that have both a fixed and a variablecomponent.

The examples in Table 3 list typical cost elements classedas fixed or variable.

These costs are always going to be a factor in establish-ing the overall cost and expense structure for a factory. Onenormally prepares a budget for a factory based on theexpected volumes for each product. This will dictate the totalbase activity for the estimating of costs for each product.

I strongly recommend that cost standards be developedfor each separate process step for each product ideally at eachisolated intermediate (although it is not necessary that eachstep be isolated). The cost standard will include all of the

Plant Operations 297

Page 313: Active Pharmaceutical Ingredients

above items, where the variable costs will be essentially thesame regardless of volume and the portion of the fixed coststhat each step will absorb.

The basis for dividing the total fixed costs among pro-ducts can be done in any way as long as the total factory fixedoverhead is fully accounted for. Some common bases are tolink the fixed overhead portion to the equipment use time orthe direct labor use.

This permits the establishing of cost standards that areuseful for production cost control.

The primary variables that should be measured againstthe ‘‘cost standards’’ as variances are:

� quantity of product (or intermediate) actually made inthe step (‘‘yield variance’’);

� quantity of auxiliary materials used in the step(‘‘charge variance’’);

� quantity of labor used for the step (‘‘labor use var-iance’’);

� unit cost of labor (‘‘labor rate variance’’);� the number of batches made in the measured time

period compared to the basis on which the standardwas established (‘‘volume variance’’), which results

Table 3 Fixed or Variable Cost Elements

Item Fixed Variable

Raw materials None AllDirect labor None AllCleaning chemicals All NoneSteam Part PartElectricity Mostly SomeWater Some MostlyWaste disposal Mostly SomeQuality testing Part PartQuality assurance All NoneSupplies Mostly SomeWarehousing All NoneDispensing None All

298 Nusim

Page 314: Active Pharmaceutical Ingredients

in a different absorption of fixed overhead thanexpected;

� the actual costs of fixed overhead that shared by theproducts is different from that originally budgeted(‘‘spending variance’’)

The pharmaceutical plant has an added cost not commonto most other chemical factories. This is the need to carry outdetailed and extensive cleaning whenever nondedicated facil-ities are used. These ‘‘cleanouts,’’ often including detailed andlaborious methods, materials, and specific testing ought to bethemselves the subject of separate cost standards.

There are a number of reasons for the establishment anduse of cost standards:

� first, to provide a direct and accurate measure of theperformance and real cost of each process step in thefactory;

� second, to provide management with a tool to measurethe effectiveness of an operation and an operatinggroup; and

� lastly, to allow the process development people apowerful tool to predict and measure the value of pro-posed improvements in the cost of manufacturing ofspecific products.

This allows management to be able to better judge thevalue of a process improvement to be sure that it is worthundertaking.

VIII. FIXED OVERHEAD ABSORPTION

In a facility dedicated to a single product, there is no questionthat product, regardless of product volume, must absorb allthe fixed overhead costs. Obviously, as the product volumeraises the unit cost of the fixed overhead absorption falls.

In a multiproduct facility this issue is less clear. All ofthe fixed overhead must still be fully absorbed; however, themechanism can vary and must be decided upon by manage-ment. One such commonly used method is to determine the

Plant Operations 299

Page 315: Active Pharmaceutical Ingredients

total labor cost planned for the year in that factory and dividethat into the total fixed overhead absorption required for sucha factory. Thus, based on the plan for the year each productindependent of the others has a specific total overhead burdento absorb (linked by standard labor used). If the quantity pro-duced is less than plan, then the product will not cover theentire fixed overhead it was scheduled to cover and generatean unfavorable variance without adversely affecting otherproducts in the factory.

In the paragraph above, it was shown how in a multipro-duct facility the impact of one product not meeting its volumeprojected in the plan does not adversely affect the other pro-ducts. However, it can have an effect on the cost of the otherproducts in the following year’s plan if the reduced require-ments for that product are not made up for by increasedvolumes of the other products it shared the facility with.

Even if the fixed overhead remained the same for thefollowing year, there would be fewer total units of productsmade at the facility forcing all of the products to absorb moreoverhead, thereby raising their apparent costs.

Much of what has been discussed in this section is notunique to pharmachemical operations but a rational approachto quantifying the real cost of a manufacturing activity.

IX. SAFETY

Again, this section deals with an issue largely the same aswith any chemical plant. It is not my intent to try to coverin detail the specifics of the subject but to focus on the generalissue of safety and point to where differences could occur as aresult of the factory making pharmachemicals.

Safety covers a variety of issues but can be separated intotwo domains: safety as it relates to personnel and safety as itrelates to facilities. Broadly speaking, pharmachemical manu-facturing because of the economics of the product can afford todeal with exotic materials and chemical process techniquesnormally too expensive to use in commodity products. Thisleads to processing techniques such as Grignard, phosgenation

300 Nusim

Page 316: Active Pharmaceutical Ingredients

even cyanide reactions. These all require handling of extre-mely hazardous materials which will require both specialfacilities to assure that the workers are protected from expo-sure and that the environment is similarly protected withdecontamination systems and controlled environment.

A. Facilities Protection

The first issue is the process itself. What fire and=or explosionhazard are inherent in the process? A review of the processwill point to obvious issues for fire or explosion such as:

� Hazardous materials:

1. liquids (acetone, alcohols, etc.) that are flam-mable and or explosion hazards;

2. solids such as sodium, sodium hydride, magne-sium, acetylenes, etc.;

3. gases such as phosgene or hydrogen cyanide.

Theuse of these kinds ofmaterialswill require close atten-tion to design to assure explosion protection is provided eitherby eliminating oxygen by inert blanketing to protect the facil-ities and perhaps remote operation to protect the personnel.

� Process:

1. The manufacturing process may allow a step topass through the explosion limits for the solventused in a given step.

2. A process step allows the concentration of anintermediate to an oil in order to eliminate thesolvent; however, continuing heating may leadto an explosion potential. This same event couldoccur unintentionally:

As an example, a common step in organic processing ismaintaining a reaction mix at a fixed temperature over a per-iod of time by refluxing the solvent from the batch. Thisrequires the solvent vapors to be condensed efficiently andredirected back to the batch to retain batch volume andconcentration. However, if some part of the reflux procedure

Plant Operations 301

Page 317: Active Pharmaceutical Ingredients

malfunctions (the cooling medium to the condenser is notavailable or the return from the condenser to the reactormisdirected or blocked) then the batch will unintentionallyconcentrate to the point where all of the solvent is gone anda concentrated oil of the reaction mass results. In many cases,this mass has been known to be a potential explosive mixture.

It is judicious, in the development process, when theprocess is finalized to examine each step for hazard potentialof these types and to add safeguards in the design to assurethat unintended hazardous conditions are either avoided orplanned for in the design.

B. Protection of Employees

1. As described above in the hazardous materials sec-tion, designing facilities to handle potentially explo-sive materials with remote operations or inertblanketing is a key factor in personnel protection.

2. Wearing respirators, air masks, or even full-sealedsuits to protect when very noxious materials areused or generated that could result in exposure tothe personnel.

Here, one must have sufficient toxicity data to know thenature of the hazard potential to personnel. Planning forthe operation includes both preparing for normal operation,which should limit exposure, and for potential deviationsin operation that could cause greater exposure. It is essentialto be overly cautious when planning for these types ofoperations.

A fact that has added greatly to this issue is theincreased potency that newer APIs have shown. This is partlyoffset in the pharmaceutical dosage form, as very smallamounts of API are required in its pill. However, the active,generated in pure form at the pharmachemical manufacturerpresents an additional challenge in processing, sampling,and testing operations at that site. It also adds a challenge tothe pharmaceutical finished dosage form manufacturer whomust handle and charge this material in a pure form to theirprocess.

302 Nusim

Page 318: Active Pharmaceutical Ingredients

X. ENVIRONMENTAL

The environmental issues faced by the pharmachemicalmanufacturer are no different from that with other fineorganic chemical producers. The concerns cover all three pos-sible routes of environmental contamination; liquid waste tosewers, solid waste generated at the site, and air pollution.

In the United States and now elsewhere, environmentalimpact statements are required to be filed before commercialprocesses can be utilized. Even after approval, the processoperation is subject to being checked periodically by a varietyof local, state, and federal review bodies.

In the United States, an environmental impact state-ment must be filed as part of the original New Drug Applica-tion. This must define the controls put in place to handle allthree possible routes of contamination.

Usually, liquid waste must meet the local standards forgoing to municipal waste treatment facility. If it doesnot meet that standard, the best resolution would be anonsite waste treatment facility. This could either be a sepa-rate pretreatment step that destroys the contaminant thatmakes the waste stream compatible with existing wastetreatment systems or definitive destruction using an onsiteincinerator.

Attempting to ‘‘ship’’ the liquid waste outside to anapproved handler is theoretically possible. However, it raisesissues of containment over the road as the waste travels to theoutside site. Concerns of leakage or an over the road accidentthat generates a spill raises all sorts of concerns that willrequire very careful planning and costly execution. It alsoassumes that plan is agreed to by all of the various environ-mental review bodies that must approve such a plan.

Solid waste generated within the processing can be apotential hazard. Again the best solution is decontaminationon site or destruction on site with an incinerator that achievesa sufficiently high temperature to assure (proven throughtesting) that the noxious materials are destroyed.

Shipping solid waste to an outside commercial facilityis more acceptable than liquid waste because of the lesser

Plant Operations 303

Page 319: Active Pharmaceutical Ingredients

issue of leakage and containment over the road. However,appropriate approvals are also needed.

Potentially contaminated gaseous materials that escapefrom the process facility must be controlled at source and onsite, scrubbing towers through which all vapor effluent fromthe process are commonly used. The preference would be todestroy the contaminant and convert it to something harm-less. Subjecting them to strong acidic or basic conditions orother known materials that are specifically reactive withthe contaminant can destroy many organics. Scrubbing iseasily set up to achieve this goal.

If the scrubbing system is only capable of capturing thematerial but not destroying it then the problem shifts todisposing of the scrubbing solution. Then the containmentand destruction shifts to the liquid steam generated by thescrubbing system.

304 Nusim

Page 320: Active Pharmaceutical Ingredients

9

Materials ManagementVICTOR CATALANO

Purchasing Group Inc. (PGI), Nutley, New Jersey, U.S.A.

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . 305II. Production Planning . . . . . . . . . . . . . . . . . . . . . 306III. Inventory Management . . . . . . . . . . . . . . . . . . . 307IV. Purchasing=Supply Management . . . . . . . . . . . . . . 308V. Distribution=Transportation . . . . . . . . . . . . . . . . . 315VI. Information Technology . . . . . . . . . . . . . . . . . . . 316VII. Quality Management . . . . . . . . . . . . . . . . . . . . 317

References . . . . . . . . . . . . . . . . . . . . . . . . . . . 318

I. INTRODUCTION

An introduction to materials management can fill an entiretextbook. The purpose of this chapter is to introduce the mostimportant aspects of materials management and for each topicto address the unique issues of the pharmaceutical industry.References are provided for those who want more detailedinformation about materials management in general andspecific areas in particular such as materials requirementsplanning.

Table 1 is a materials management matrix. It shows thata primary objective of marketing is to increase customer ser-vice, of manufacturing to decrease operating expenses, and offinance to decrease inventory. It also shows that as marketingtries to increase customer service, there is a tendency for

305

Page 321: Active Pharmaceutical Ingredients

operating expenses and inventory to increase contrary to theobjectives of manufacturing and finance. Likewise as manu-facturing tries to decrease operating expenses, there is a ten-dency for customer service to decrease and inventory toincrease. As finance tries to decrease inventory, there is a ten-dency for customer service to decrease and operatingexpenses to increase.

Therefore, there is a need to balance conflicting objectives.Materials management requires that conflicting objectives bebalanced. The best approach to materials management (andto most aspects of life) is a balanced approach.

II. PRODUCTION PLANNING

The objective of production planning is to coordinate the useof a company’s resources (materials, processes, equipment,and labor) to make the right goods at the right quality atthe right time at the right (lowest total) cost.

The right quality is key in the pharmaceutical industry.Equipment utilized may be dedicated to a single product or bemultipurpose. The multipurpose equipment requires thor-ough clean out between different products. This clean outand turnaround time between different products can be sig-nificant. A balance must be made between longer productionruns (fewer clean outs) and generally higher inventory andshorter production runs (more clean outs) and generally lowerinventory.

In the pharmaceutical industry, sometimes it makesmore sense to have several dedicated lines, rather than one

Table 1 Materials Management Matrix

Customer service Operating expenses Inventory

Marketing Increase Increase IncreaseManufacturing Decrease Decrease IncreaseFinance Decrease Decrease Decrease

306 Catalano

Page 322: Active Pharmaceutical Ingredients

multipurpose line to avoid the cost of clean outs andturnaround.

Production planning is made more challenging in thepharmaceutical industry with the introduction of newproducts. Trying to forecast the amount and timing for newproducts is made more difficult because of the need for phar-maceutical products to be approved by the Food and DrugAdministration (FDA) before the products are allowed to besold.

Pharmaceutical companies usually have to provide capa-city for the new product before the new product is approved.Sometimes, this initial production capacity may be outsourcedto a custom manufacturer. In fact, outsourcing has becomemore important to the pharmaceutical industry as pharma-ceutical companies focus internal resources on developingnew products, and take advantage of the manufacturing capa-city of external sources for both initial and also long-termproduction capacity.

III. INVENTORY MANAGEMENT

Nowhere is a balanced approach more important than ininventory management.

I still remember the advice given to me by my managerwhen I first began purchasing materials for manufacturing.My manager explained that inventories should be kept aslow as possible. My manager further explained that if inven-tories were found to be too high I would probably be given a‘‘slap on the hand.’’ But my manager warned that if inven-tories should ever drop so low that the manufacturing opera-tion was interrupted or a plant shut down, I would probablylose my job. This advice made it clear to me which way Ishould err—the manufacturing operation was never shutdown because of lack of production materials.

What is unique about the pharmaceutical industry isthat the finished product is a product that can literally meanthe difference between life and death for a patient in need of alife saving drug. To run out of such a product could be deadly.

Materials Management 307

Page 323: Active Pharmaceutical Ingredients

Another reason why inventories might be maintained athigher levels in the pharmaceutical industry compared toother industries is that the pharmaceutical industry has his-torically been more profitable than many other industries. If asale is lost because of lack of inventory, the opportunity cost issignificant. The cost of the finished good is relatively smallcompared to the selling price of the pharmaceutical product.

It is interesting to note that even the U.S. Governmentkept inventory of pharmaceutical products, such as morphine,in its strategic warehouses to be prepared in the event of waror other situations where these important products might beneeded.

The three primary categories of inventory to be managedare raw materials, work in process (WIP), and finished goods.

When materials are received to be used in the manufac-ture of a pharmaceutical product, an inspection (and possibletesting) is usually necessary before the materials are accepted.Materials may be stored in a separate ‘‘quarantined’’ areauntil they are released and approved for use. The use of certi-fied suppliers has allowed some materials to be received andaccepted without additional inspection or testing. The certifiedsupplier may provide a ‘‘certificate of analysis’’ documentingthat the material meets the required specifications. Certifiedsuppliers are usually audited to insure that they meet therequirements for certification of the customer.

IV. PURCHASING=SUPPLY MANAGEMENT

Purchasing=procurement=supply management=supply chainmanagement are some of the names that are used to describea function that continues to grow in value to organizations ingeneral and pharmaceutical companies in particular.

Purchasing involves obtaining the right material (or ser-vices), at the right quality in the right quantities, at the righttime, from the right source, at the right price.

Again the right quality is significant to the pharmaceuti-cal industry. The supplier must understand good manufactur-ing practice (GMP). Supplier selection is critical. Supplier

308 Catalano

Page 324: Active Pharmaceutical Ingredients

certification is important to many pharmaceutical companies.Suppliers must understand the importance of meeting speci-fications and controlling their processes to meet specificationsconsistently. Suppliers must notify the pharmaceuticalcompany of any process changes.

Organizations generally spend a significant portion oftheir sales dollar in the purchase of materials and supplies,and an even greater share when services and capital areincluded. A small reduction in purchase cost may have thesame impact on profit as a much larger increase in sales. Thistremendous potential to increase profits is what has causedmany organizations to focus more attention on the importanceof purchasing and supply management. Purchasing and sup-ply management professionals are adding value to theirorganizations through strategic cost reduction.

Some of the cost reduction strategies that have been usedinclude:

i. Supplier consolidation: Organizations are redu-cing the number of suppliers that they do busi-ness with. Reducing the number of suppliersfor a particular commodity increases leverage.You become a more important customer to thesupplier. The supplier can reduce the pricebased on the increased volume. Dealing withfewer suppliers allows for better supplier man-agement. Purchasing and supply managementprofessionals have limited time, and can onlyeffectively work with a limited number of suppli-ers. Consolidating suppliers allows more time tobe spent with the most important suppliers.

ii. Specifications: Specifications are important incost reduction and management. You shouldpurchase only what you need and not over spe-cify. As an example when purchasing a chemical,the chemical may be offered in several gradeswith different levels of purity such as 98% or99%. It is a waste to purchase the more expen-sive 99% purity chemical if the 98% purity

Materials Management 309

Page 325: Active Pharmaceutical Ingredients

chemical will meet the requirements to producethe desired product.

iii. Standardization: Standardization helps reducethe number of different items purchased. Forexample in the area of office supplies, insteadof buying 10 different pens, purchase only threedifferent pens. This increases the volume of thethree selected pens and reduces the price andcost. For process equipment such as pumps,instead of purchasing 10 different pumps, pur-chase only three different pumps. This increasesthe volume of the three selected pumps andreduces the price and cost. But this also resultsin other benefits such as ease of operation andmaintenance for operators and mechanics whobecome more familiar with the three selectedpumps. Another benefit is the reduction of spareparts. If you have 10 different models of pumpsyou will need spare parts for all 10 models. Ifthere are only three different models, you needto hold spares for only the three differentmodels.

iv. Competitive bidding: Organizations can reducecosts through competitive bidding. This requiresgood specifications and a good scope of work.Purchasing will develop a request for proposal(RFP) that is sent to the preselected and quali-fied suppliers. Today many of these RFPs aresent over the Internet as electronic request forproposals or eRFPs. There is also an increaseduse of reverse auctions, where preselected andqualified suppliers will bid down the price of aspecific commodity with the business awardedto the qualified supplier with the lowest bid.

v. Negotiation: Negotiation is a key tool used toreduce cost. Good negotiation requires good pre-paration. Developing a negotiation brief beforethe negotiation is helpful in preparation for asuccessful negotiation. The negotiation brief will

310 Catalano

Page 326: Active Pharmaceutical Ingredients

include information such as the history of thesuppliers—current and potential and pricing. Itwill list the objectives of the negotiation includ-ing what must be achieved and what would benice to have. Negotiation is a skill that can begained through education (courses such asKarrass) and experience. Successful negotiationsare usually positive and are a win–win for boththe supplier and the customer. It is usuallypossible to find a better deal for both parties.

vi. Make vs. buy: Many pharmaceutical companieshave chemical manufacturing capability. Whena new product is being developed these pharma-ceutical companies have the option to manufac-ture the chemical or to purchase it from anoutside chemical supplier. This allows the phar-maceutical company to do the make vs. buy ana-lysis and select the lowest cost alternative.During times when there is much external che-mical manufacturing capacity, chemical suppli-ers are willing to ‘‘sharpen their pencils’’ andoffer attractive pricing for chemicals.

vii. Outsourcing: Organizations should decide whatbusiness they are in and what is their corecompetency. This is true for pharmaceuticalcompanies. Pharmaceutical companies may beinvolved with activities that are not core. As anexample, facilities maintenance is an area thatis not core that could be considered for outsour-cing. To outsource successfully, it is importantto develop the scope of the work that is to be out-sourced. It is important to include all that is tobe outsourced. If you miss something the organi-zation doing the outsourced work will be glad toadd what you missed, but usually at a muchhigher price than if it had been included in theoriginal scope of work. To make sure to includeall that you want to outsource, it is importantto have an outsourcing teamwith all the involved

Materials Management 311

Page 327: Active Pharmaceutical Ingredients

parties and especially those who are the expertsin what is to be outsourced. The outsourcingteam should also include a representative fromHuman Resources as some employees may beoutsourced.

viii. Policy: An organization’s policy can have animpact on costs. A good example is a companypolicy on travel. Many companies have a policyon the class of air travel. A company may notallow and may not reimburse employees to tra-vel by air first class. Business class travel maybe allowed only for trips longer than a specifiedduration such as longer than 8 hr. All other tra-vel should normally be economy. How the policyis set could help save the company significantly,but the policy must balance the needs of the tra-veling employee with the associated costs. If onlyeconomy class air travel is allowed, and employ-ees are required to travel frequently on longflights such as betweenNewYork and Singapore,the employees may arrive tired and not be as pro-ductive as if they had been allowed to travel onbusiness class.

ix. Long-term agreements: Long-term agreements(3 years or longer) generally result in cost reduc-tion. Suppliers are more willing to offer betterpricing and invest more time to help with costsaving ideas. The relationship between supplierand customer becomes more of an alliancerelationship.

x. Global sourcing: Globalization has affected allorganizations and pharmaceutical companies inparticular. Cost savings can be achieved byselecting the best suppliers in the world. Manyof the chemicals and other materials that areused in the manufacture of pharmaceuticals aremanufactured around the world in such coun-tries as England, France, Germany, Switzerland,Italy, and Japan. In the future, more will be

312 Catalano

Page 328: Active Pharmaceutical Ingredients

manufactured in emerging markets such asChina and India. And the quality from thesemanufacturers can be excellent. When there aremultiple suppliers on more than one continentfor a particular chemical or material, purchasingcan take advantage of exchange rates to obtainbetter pricing and reduce total costs.

xi. Reduce freight costs: Reducing freight costs ofthe incoming raw materials and supplies andthe outgoing final products is an important wayto reduce overall costs. There are opportunitiesfor savings in all of the various modes of trans-portation including truckload, less than truck-load (LTL), air, rail, ocean, and small package.

xii. Reduce lead time: Reducing lead times is espe-cially important to pharmaceutical companieswhen introducing new products. This allowsorganizations to obtain materials needed quicklyfrom suppliers to meet unanticipated demand.Reduced lead times improves the time to marketa new product.

xiii. Reduce inventory: There is a need for balancewhen reducing inventory. Inventory should bekept low to reduce the dollars tied up in the costof the inventory, and to reduce the carrying costsof the inventory, and to reduce the cost of inven-tory, which may go bad or expire. But inventorymust be kept high enough to keep operationsrunning smoothly without interruption. Theinventory of final product should be kept highenough to make sure that every customer thatneeds the product can get it. For pharmaceuticalproducts this is especially important since theseproducts could mean the difference between lifeand death for the patient.

xiv. Reduce demand: Reducing demand is one of thebest ways to reduce cost. If you are able tonegotiate a 15% discount on an item, you cansave 15%. But if you can find a way to eliminate

Materials Management 313

Page 329: Active Pharmaceutical Ingredients

the need to purchase that item, you have saved100%—the full cost of the item. An example isa company that changed its travel policy to elim-inate all business class air travel. Prior to thepolicy change an employee could travel betweenNew York and Singapore on business class. Oneparticular traveler averaged four trips betweenNew York and Singapore a year. After the policychange this traveler saved much more than thedifference between the price of the businessand economy class fare, because this travelermade only two trips instead of four trips. Forthe two trips that were not taken the travelersaved the company the full fare.

xv. Reengineer purchasing process: Reengineeringcan start by documenting or mapping the cur-rent process. The next step is to identify andeliminate the nonvalue adding or unnecessarysteps. Most employees are being asked to domore with less, so it is important to examineall that is being done, and eliminate the nonva-lue adding activities. Another approach to reen-gineering is to start with a blank piece of paperand list only the value adding activities thatshould be done.

xvi. Automation=information technology: It is impor-tant to reengineer first so that the process that isautomated is a good one. If you automate a ‘‘bad’’process you will just do the ‘‘bad’’ process quickerand consistently. Many companies haveinstalled enterprise-wide information resourcesystems such as Oracle and SAP. These allowbetter information to be accessed for better deci-sion making. It allows supply managers to seeand better understand the spending—how muchof what is being purchased by which suppliers.There are other systems that are being imple-mented to help direct the purchasing to the pre-ferred suppliers to take advantage of the cost

314 Catalano

Page 330: Active Pharmaceutical Ingredients

saving agreements that have been negotiated.Many of these transactions are taking place orwill take place in the future across the Internet.

xvii. Transfer best practices: To improve perfor-mance, it is important to identify best practicesand to implement them. If a best practice isfound within an organization, it should beapplied across the entire organization. Best prac-tices should also be identified in other organiza-tions and should be adopted (steal from thebest). There are organizations such as the Insti-tute for Supply Management (ISM) and the DrugChemical and Allied Trades Association (DCAT)that offer seminars and programs to learn aboutthese best practices. Within ISM there is thePharmaceutical Forum and the Chemical Group,which along with DCAT offer educational andnetworking opportunities.

xviii. Energy conservation: Energy conservation isand will continue to be an important way toreduce demand and reduce costs.

xix. Measurements: Measurements are needed toidentify the opportunities for savings. Measure-ments are also used to improve performanceinternally and externally the performance ofsuppliers. It is important to measure what youwant to manage.

V. DISTRIBUTION=TRANSPORTATION

Distribution involves getting the product to the customer atthe right time. Channels are the particular paths in whichthe goods move through distribution centers, wholesalers,and retailers.

Distribution requirements planning is a systemapproach that allows for distribution at minimum total cost.

Transportation involves the movement of raw materialsfrom suppliers to production and finished goods to customers.

Materials Management 315

Page 331: Active Pharmaceutical Ingredients

Transportation involves a variety of modes including air, rail,motor freight, truckload (TL) and less than truckload (LTL),and ocean freight.

Many pharmaceutical products must be maintainedwithin specific temperature ranges. Some of these productsmust be shipped in refrigerated containers or trailerscommonly referred to as ‘‘reefers’’ at a premium price.

With many pharmaceutical companies having facilitiesin Puerto Rico (because of the tax advantages), ocean ship-ments between the United States and Puerto Rico are com-mon. Again many of these ocean shipments are made inrefrigerated or temperature-controlled containers.

Even though air transportation is usually the most costlyform of transportation, some pharmaceutical products areshipped by air for reasons of timing.

The pharmaceutical industry is a global industry. Sup-pliers are selected from the best suppliers in the world and cus-tomers are located worldwide. Therefore, raw materials andfinal products are shipped all over the world, making transpor-tation an important function within materials management.

VI. INFORMATION TECHNOLOGY

Information technology (IT) is an important area and will con-tinue to become more important as time goes on. Many com-panies including pharmaceutical companies have been, orare in, the process of implementing enterprise informationsystems. For a global company to be able to pull togetherinformation on a global basis is of great value. For example,to have access to the inventory of a raw material or a finishedgood quickly on a global basis has allowed companies toreduce the amount of inventory. There is a substitution ofinformation for inventory. The better the information avail-able on inventory the lower the levels of inventory requiredto satisfy customer requirements. The better the informationavailable on what has been and is to be purchased fromsuppliers the greater the negotiating leverage with thosesuppliers.

316 Catalano

Page 332: Active Pharmaceutical Ingredients

The particular system selected may not be as important asthe need to understand if the systemmeets the requirements ofthe company. It is also better if the company implements anenterprise wide system consistently across the entire company.

Some companies have selected information technologyproducts with a ‘‘best of breed’’ approach. They selected thebest purchasing system, the best accounts payable system,the best manufacturing resource planning system, etc. In the-ory, this should yield the best overall system. In fact, severalpharmaceutical companies that implemented this ‘‘best ofbreed’’ approach now realize some of the disadvantages of try-ing to interface the various systems. Each time anyone of theindividual systems is upgraded, there is the difficulty ofupgrading the interface between the various systems.

Several of these companies now suggest that the ‘‘best ofbreed’’ approach may not be the best and that one overallsystem may be a better approach.

Many pharmaceutical companies are now using theInternet to communicate with and transact business withsuppliers and customers. The use of the Internet by pharma-ceutical companies will continue to grow.

VII. QUALITY MANAGEMENT

Quality management has become important to almost everyindustry, but remains even more important to the pharma-ceutical industry.

I have attempted to provide examples of the importanceof quality in many of the previous sections of this chapter.Quality is so important to the pharmaceutical industry thata complete chapter of this text has been devoted to quality.

Some of the areas to consider with regards to qualityinclude current good manufacturing practices (cGMP), ISO9000 requirements, auditing, and validation.

Current good manufacturing practices provide the mini-mum guidelines for the production of drugs that are safe,pure, and effective. The Food and Drug Administration ischarged with enforcing all provisions of the Food, Drug, and

Materials Management 317

Page 333: Active Pharmaceutical Ingredients

Cosmetic Act and regulations. The cGMPs are part of theseregulations.

ISO 9000 in an international standard for qualitymanagement systems. The standards are not specific forany particular industry, but have been adopted and are usedat least by some pharmaceutical companies. ISO exists as aseries of standards that covers design and development, pro-duction, installation and servicing, and inspection. ISOrequires that material identification and traceability be main-tained, that suppliers are evaluated on a regular basis, andthat training programs are established and documented.

Audits are conducted as a management tool for assessingthe quality level of an operation. They are used to identify non-conformance and tomake corrective actions as needed, and pre-vent reoccurrence of potential problems that can adverselyaffect a product. Audits are conducted internally and externally.Supplier audits may be directed ‘‘for cause’’, such as a customercomplaint, for change control, or for a product problem. Auditsmay be scheduled on a regular basis (e.g., every 3 years) for sup-pliers of key or critical materials.

REFERENCES

The references are provided for those who desire moredetailed information about Materials management in generaland specific areas in particular.

1. Adams ND. Warehouse and Distribution Automation Hand-book. 1996.

2. Allegri .H. Materials Management Handbook. 1991.

3. Anderson BV. The Art and Science of Computer AssistedOrdering: Methods for Management. 1996.

4. Arnold JRT. Introduction to Materials Management. 1998.

5. Ashley JM. International Purchasing Handbook. 1998.

6. Baker RJ. Policy and Procedures Manual for Purchasing andMaterials Control. 1992.

7. Ballou RH. Business Logistics Management. 1991.

318 Catalano

Page 334: Active Pharmaceutical Ingredients

8. Bigelow CR. Hazardous Materials Management in PhysicalDistribution. 1997.

9. Bolten EF. Managing Time and Space in the Modern Ware-house: with Ready-to-Use Forms, Checklists & Documentation.1997.

10. Bowersox DJ, Closs DJ. Logistical Management: the Inte-grated Supply Chain Process. McGraw-Hill Series in Market-ing. 1996.

11. Burgess WA. Recognition of Health Hazards in Industry: aReview of Materials and Processes. 1995.

12. Carter JR. Purchasing: Continued Improvement ThroughIntegration. Business One Irwin=Apics Library of IntegratedManagement. 1992.

13. Carter S. Successful Purchasing. Barron’s Business SuccessSeries. 1997.

14. Chadwick T. Strategic Supply Management: an Implementa-tion Toolkit. 1996.

15. Clement J. Manufacturing Data Structures: Building Founda-tions for Excellence With Bills of Materials and Process Infor-mation. 1995.

16. Copacino WC. Supply Chain Management: the Basics andBeyond. Apics Series on Resource Management. The St. LuciePress. 1997.

17. Dobler DW. Purchasing and Supply Management: Text andCases. McGraw-Hill Series in Management. 1995.

18. Ellram LM, Birou LM (Contributor). Purchasing for BottomLine Impact: Improving the Organization Through StrategicProcurement. The NAPM Professional Development Series.Vol. 4. 1995.

19. Farrington B (Contributor), Waters DWF. The Services Buyerin the Role of Project and Cost Management. 1998.

20. Fernandez RC. Total Quality in Purchasing & Supplier Man-agement (Total Quality). 1995.

21. Ford WO. Purchasing Management Guide to Selecting Suppli-ers. 1995.

Materials Management 319

Page 335: Active Pharmaceutical Ingredients

22. Grieco PL. MRO Purchasing. The Purchasing Excellence Ser-ies. 1997.

23. Grieco PL. Power Purchasing: Supply Management in the 21stCentury. 1995.

24. Grieco PL. Suppy Management Toolbox: How to Manage YourSuppliers. 1995.

25. Handfield RB. Introduction to Supply Chain Management.1998.

26. Harmon RL. Reinventing the Warehouse: World Class Distri-bution Logistics. 1993.

27. Hassab JC. Systems Management: People, Computers,Machines, Materials. 1996.

28. Hickman TK. Global Purchasing: How to Buy Goods andServices in Foreign Markets. Business One Irwin= ApicsSeries in Production Management. 1992.

29. Hough HE. Handbook of Buying and Purchasing Management.1992.

30. Killen KH. Managing Purchasing: Making the Supply TeamWork. NAPM Professional Development. Vol. 2. 1995.

31. King DB. Purchasing Manager’s Desk Book of PurchasingLaw. 1997.

32. Krotseng L. Global Sourcing. The Purchasing ExcellenceSeries. 1997.

33. Lambert DM. Fundamentals of Logistics Management. TheIrwin= McGraw-Hill Series in Marketing. 1997.

34. Lambert DM. Strategic Logistics Management. Irwin Series inMarketing. 1992.

35. Laseter TM. Balanced Sourcing: Cooperation and Competitionin Supplier Relationships. 1998.

36. Leenders MR. Value-Driven Purchasing: Managing the KeySteps in the Acquisition Process. The NAPM ProfessionalDevelopment. Vol. 1. 1994.

37. Leenders MR. Purchasing and Materials Management. 1992.

320 Catalano

Page 336: Active Pharmaceutical Ingredients

38. Locke D, Locke R. Global Supply Management: a Guide toInternational Purchasing. NAPM Professional DevelopmentSeries. 1996.

39. Lunn T. MRP: Integrating Material Requirements Planningand Modern Business. Business One Irwin= Apics Series inProduction Management. 1992.

40. Mulcahy DE. Materials Handling Handbook 1998.

41. Narasimhan SL. Production Planning and Inventory Control.Quantitative Methods and Applied Statistics Series. 1995.

42. Newman RG. Capital Equipment Buying Handbook. 1998.

43. Orlicky J. Orlicky’s Material Requirements Planning. 1994.

44. Pilachowski M. Purchasing Performance Measurements: aRoadmap for Excellence. Purchasing Excellence Series. 1996.

45. Poirier CC. Supply Chain Optimization: Building the StrongesTotal Business Network. 1996.

46. Pooler VH. Global Purchasing: Reaching for the World. VNRMaterials Management= Logistics Series. 1992.

47. Pooler VH. Purchasing and Supply Management: Creating theVision. Materials Management=Logistics Series. 1997.

48. Ptak CA. MRP and Beyond: a Toolbox for Integrating Peopleand Systems. 1996.

49. Raedels AR. Value-Focused Supply Management: Getting theMost Out of the Supply Function. The NAPM ProfessionalDevelopment Series. Vol. 3. 1994.

50. Riggs DA, Robbins SL (Contributor). The Executive’s Guide toSupply Management Strategies: Building Supply ChainThinking into All Business Processes. 1998.

51. Robeson JF (Preface), Copacino WC (Editor). The LogisticsHandbook. 1994.

52. Romme J (Editor), Hoekstra SJ. Integral Logistic Structures:Developing Customer-Oriented Goods Flow. 1992.

53. Ross DR. Distribution: Planning and Control. Chapman & HallMaterials Management=Logistics Series. 1995.

Materials Management 321

Page 337: Active Pharmaceutical Ingredients

54. Scheuing EE, Scheuing E. Value-Added Purchasing: Partner-ing for World-Class Performance. Crisp Management Library.1998.

55. Steele PT, Court B (Contributor). Profitable Purchasing Stra-tegies: a Manager’s Guide for Improving Organizational Com-petitiveness Through the Skills of Purchasing. 1996.

56. Tersine RJ. Principles of Inventory and Materials Manage-ment. 1994.

57. Underhill T. Strategic Alliances: Managing the Supply Chain.1996.

58. Van Mieghem T, Mieghem TV. Implementing Supplier Part-nerships: How to Lower Costs and Improve Service. 1995.

59. Wood DF. International Logistics. Chapman & Hall MaterialsManagement=Logistics. 1994.

60. Woodside G. Hazardous Materials and Hazardous Waste Man-agement: a Technical Guide. 1993.

61. Zenz GJ, Thompson GH (Editor). Purchasing and the Manage-ment of Materials. 1993.

322 Catalano

Page 338: Active Pharmaceutical Ingredients

10

Plant MaintenanceRAYMOND J. OLIVERSON

HSB Reliability Technologies, Kingwood, Texas, U.S.A.

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . 323II. Strategic Plan . . . . . . . . . . . . . . . . . . . . . . . . . 324III. Reliability-Balanced Scorecards . . . . . . . . . . . . . . 325IV. Maintenance Basics . . . . . . . . . . . . . . . . . . . . . 327V. Condition Monitoring . . . . . . . . . . . . . . . . . . . . 329VI. Operator-Driven Reliability . . . . . . . . . . . . . . . . . 330VII. Reliability Engineering . . . . . . . . . . . . . . . . . . . . 331VIII. Risk Management . . . . . . . . . . . . . . . . . . . . . . 333IX. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . 334

I. INTRODUCTION

Pharmaceutical plants need to address maintenance andreliability issues better in order to produce life critical pro-ducts, on time, on cost, and on quality, safely with respectfor personnel, property, and the environment. Also, thereare issues of evidence of service and other FDA, OSHA, andEPA compliance factors that impact the maintenance func-tion in pharmaceutical plants. These issues are discussed inother chapters within this book.

Our experience in pharmaceuticals shows that plantspursuing manufacturing excellence develop a strategicplan for maintenance and reliability and are focused onreliability-balanced scorecards, maintenance basics, condition

323

Page 339: Active Pharmaceutical Ingredients

monitoring, operator-driven reliability, reliability engineer-ing, and risk management. These issues are basically the samefor continuous process plants or batch plants. However, it hasbeen our experience that a sound grasp of maintenance excel-lence is lacking within the typical pharmaceutical batch typeplant. There seems to be a stronger focus on launching newproducts and meeting production schedules than on day-to-day maintenance.

II. STRATEGIC PLAN

Successful pharmaceutical plants develop a multiyear, strate-gic plan for maintenance and reliability. The key issues aredeveloped with a cross-section of plant personnel. A properstrategic plan will involve the following issues:

A. Vision: Where will we be in 3–5 years?B. Mission: How will the vision be reached?C. Goals and objectives: How will we know when we

get there and what it was worth?D. Philosophy: What will our maintenance and relia-

bility culture be?E. Organization structure: How will our structure

change during the journey?F. Rewards: How will all personnel be recognized for

their efforts and achievements, ideally in a groupsense?

G. Training: What training will be required to reachour destination?

H. Maintenance role: How will the role of the mainte-nance department change during the next 3–5years? Will the maintenance department exist asa separate function in 5 years?

I. Technology: What role will technology play? Anexample is interfacing process control computersto a computerized maintenance management sys-tem (CMMS) for integrated condition monitoring,which will lead to a sound predictive maintenanceprogram.

324 Oliverson

Page 340: Active Pharmaceutical Ingredients

J. Capital strategy: What sort of capital spending willbe required to reach our goals?

K. Work force strategy: How will hourly personnel beinvolved? Will we have to negotiate significantchange?

L. Customer strategy: How will our customers beinvolved in our journey? What will our achieve-ments mean to our customers?

M. Vendor strategy: What will change in our relation-ships with our vendors of services and materials,and how will the benefits be measured?

III. RELIABILITY-BALANCED SCORECARDS

Once a strategic plan is in place, pharmaceutical companiesneed to implement a reliability-balanced scorecard (RBS) tointegrate measures to achieve their vision and mission. TheRBS tracks performance and measures results across four dis-tinct areas—financials, business processes, innovations andlearning, and customer support. Historically, companies havefocused on only one track of measurements. For example,from a shareholder’s viewpoint, the focus was always finan-cials. If manufacturing became the focus, then units producedto plan would be a measure. With the RBS, companies havethe opportunity to integrate the lagging indicators of finan-cials and customer support with the leading indicators ofbusiness processes and innovations and learning.

The RBS provides an integrated set of measurementsfrom unit level to plant level to division level to corporate level.For the first time, the company can tie their budgeting withtheir strategic plans and track month by month how theyare progressing. However, in setting up the scorecard, caremust be taken in developing the measures at each level. Phar-maceutical companies need to ensure that their measuresmeet each of the criteria in Figure 1. Performance measuresshould relate vision and mission to the strategy and tacticsestablished to meet a set of goals. Performance measuresshould not all be financial. Each unit or location can havemeasures that vary from others. They can change over time.

Plant Maintenance 325

Page 341: Active Pharmaceutical Ingredients

Figure 1 Performance measures.

For example, if one of the strategies to improve reliability is toimplement a preventive maintenance (PM) program, then thefirst set of measures would be to track the number of equip-ment items that have PMs established. This would hold untilthe PM program was in place, then the relevant measurewould change to PM completion rates. Next, the measuresshould be simple and easy to use. This is critical for accep-tance at each of the levels that the RBS touches. Performancemeasures, in all cases, should provide fast feedback. This isnecessary for two reasons as follows. As the units review theirprogress toward established goals, they need to know theimpact the change is making. A business process change thatcan have dramatic impact monthly should not be measuredquarterly. The units and management must be able to controlthe impact of change. Finally, the performance measureshould foster improvement. All measures should be used totrack and control positive change within the company andthe individuals affected (Fig. 2).

By using these principles and building a relevant set ofmeasures, pharmaceutical companies can set the plants onthe right path to reliability improvements. For example, effec-tive planning and scheduling of maintenance work can affectseveral process indicators, such as schedule compliance or per-cent of planned work. Results indicators include such items aspercent overtime or percent ‘‘E’’ work. The financial changethat can be tracked through the improvement in planning

326 Oliverson

Page 342: Active Pharmaceutical Ingredients

and scheduling of work can be measured through percentmaintenance cost to replacement asset value.

Through implementation of the RBS, pharmaceuticalcompanies have the opportunity to bring a modern approachof measuring change to their companies. Using the RBS tomeasure the improvements in maintenance and reliabilitywill provide an integrated view of reliability impact on suchareas as financial, customer support, business process, andinnovation and learning.

IV. MAINTENANCE BASICS

There are a number of issues that must be addressed in themaintenance basics arena in order for a pharmaceutical plantto achieve excellence in maintenance. They are performancemeasures, work order controls, preventive maintenance,spare parts management, operations=maintenance relations,training=continuing education, and the CMMS.

Figure 2 Performance measurement flow path.

Plant Maintenance 327

Page 343: Active Pharmaceutical Ingredients

A. Performance measures: The first step in establish-ing an effective maintenance basics program is toidentify and establish performance measures. Mostexperienced managers have learned that ‘‘you getwhat you measure.’’ Successful pharmaceuticalplants develop and implement a series of supportivekey performance measures to track and manageimprovement of maintenance=reliability. Examplesare mean time between failure, maintenance costper unit of output, percent planned and scheduledmaintenance, preventive maintenance tasks com-pleted, expense maintenance cost as a percentageof replacement asset value, etc. These indicatorsmust be available to all levels of an organizationand should be used to maximize individual andgroup contributions. It would be best if these indica-tors were part of the reliability-balanced scorecard.

B. Work order controls: We have found that the mostcost-effective plants have a systematized approachto identifying, prioritizing, planning, scheduling,executing, and recording routine maintenance. Thiswould include plant shutdowns.

C. Preventive=predictive maintenance: Cost-effectiveplants have an equipment criticality ranking schemeand use reliability-centered maintenance (RCM)techniques to determine the equipment that requirespreventive maintenance. They use a combinationof equipment manufacturers’ recommendations,experience, and available PM databases to validateor modify existing PMs and to create new preventivemaintenance schedules where necessary; then theyfaithfully execute their PM programs.

D. Spare parts (stores) management: Getting the rightspare part to the right place on time is an importantstep in effective maintenance materials manage-ment. Cost-effective pharmaceutical plants employproper systems, procedures, and practices relatingto the procurement and management of mainte-nance spare parts. Size of spare parts inventory

328 Oliverson

Page 344: Active Pharmaceutical Ingredients

and other procurement costs are often excessive inpharmaceutical plants because of the ongoing cam-paigns to produce new products with ever-changingplant equipment. Vigilance is required to managethis situation. Vendor stocking programs (VSPs),consignment inventory, electronic data interchange(EDI), bar coding, cycle counting, and other techni-ques should be used to reduce inventory levels.

E. Operations=maintenance relations: Solid customer=supplier relationships, or even better, partnershipsbetween production and maintenance groups areessential in the pursuit of maintenance excellence.The enemy is not inside the plant walls.

F. Training=continuing education: Companies mustdetermine skills training requirements and pro-vide the training. They also must focus on consis-tent implementation of roles and responsibilitieswith all levels of the organization to ensure thatthe right things get done correctly, on time, andsafely. A special subset of roles and responsibilitiesis multiskill maintenance. The pacesetter plantshave a flexible work crew with a broad base ofskills who are supported by specialists.

G. CMMS: The final factor in maintenance basics, theCMMS, ties all the issues together. Actually, it insti-tutionalizes thebehaviorsrequired toachievemainte-nance excellence. Typically, it has a work controlmodule, an inventory module, a purchasing module,and ties to financial systems, payables, general led-ger, etc. Today, there are several ‘‘on-condition’’ sys-tems such as Ivara’s EXP that enhance a CMMS. Akey issue with a CMMS or on-condition system is thatit can, if used properly, provide the ‘‘evidence of ser-vice’’ required by the FDA.

V. CONDITION MONITORING

There are three steps that a pharmaceutical plant should taketo implement an effective condition-monitoring program.

Plant Maintenance 329

Page 345: Active Pharmaceutical Ingredients

1. Identify equipment: The first step in implementinga condition-monitoring program is to deter-mine what equipment will be monitored. It willbe critical equipment and generally is about 10%of the total equipment. All manufacturing andenvironmental control equipment should be evalu-ated.

2. Select technologies: The next step is to choose thetechnologies that will be employed. Typically,well-maintained and reliable pharmaceuticalplants employ vibration monitoring, thermogra-phy, ferrography, particulate count, detailed motoranalyses, and environmental monitoring.

3. Implementation: Routes and timing are established,people are trained, and the program is implemen-ted. Readings are taken, analyses are performed,and recommendations for action are provided tomaintenance, engineering, and operations. A strictcondition-monitoring discipline will lead a plant toa successful predictive maintenance environmentwhere people at all levels of a plant will be able tovisualize the resultant equipment ‘‘saves.’’

VI. OPERATOR-DRIVEN RELIABILITY

The items listed under the category of operator-driven relia-bility are fairly self-evident. The intent is to increase opera-tors’ ownership of their equipment and its reliability. Theprocess encompasses many of the activities of total productivemaintenance (TPM). Three key areas of focus are:

1. Improving adherence to equipment procedures(start-up, operation, and shutdowns);

2. Improving communication, coordination, and pro-blem solving between production and maintenance;

3. Increasing operators’ responsibilities in housekeep-ing, equipment inspection, and performance ofminor maintenance.

330 Oliverson

Page 346: Active Pharmaceutical Ingredients

VII. RELIABILITY ENGINEERING

The area of reliability engineering finds the pharmaceuticalindustry well behind the refining=petrochemical industry. Agreat deal of ‘‘catch-up’’ will be required for the typical phar-maceutical plant. The main areas of catch-up are listed below.

A. Reliability-centered maintenance: RCM providesan engineering, risk-based technique for managingequipment performance. The method takes thelarge, highly nonintuitive problem of identifyinghigh-risk failure modes and divides it into manysmall, easily solved problems in order to design arisk-based maintenance plan.

B. Failure mode study: Potential failure modes ofcritical equipment are ranked according to theirrisk (probability � consequence). Typically, 80% ofequipment’s total risk is due to 30% of its failuremodes. The failure mode study allows managementto allocate scarce resources (labor, materials, andequipment) on a cost-effective basis to attackhigh-risk potential failures.

C. Analysis of root cause of recurring failures: In mostpharmaceutical plants, over one-half of the workorders completed are unnecessary or preventable.Plants need to establish a systematic approach ofrecognizing=analyzing recurring failures and deter-mining=correcting the root causes. This eliminatesunnecessary downtime and reduces maintenanceexpenditures for labor and materials. Also there isreduced risk of safety and environmental incidents.One way to look at unnecessary or preventable workis to review past work orders and determine whetherthey were necessary or not. By grouping them intonecessary and unnecessary categories, plants areable to determine where to focus on reducing preven-table work. Figure 3 illustrates a typical pharma-ceutical plant’s results typical plant results withnecessary work at 51% and unnecessary work cate-

Plant Maintenance 331

Page 347: Active Pharmaceutical Ingredients

gorized as operations caused work (13%), mainte-nance rework (14%), design problems (18%), andmanagement lack of support for training, preventiveand=or predictive maintenance, etc. (4%).

D. Maintainability review: This three prongedapproach to improving equipment reliability isbased on failure analysis to identify root causes,testing of equipment immediately after repair toensure quality work was performed, and perfor-mance analysis of equipment to determine equip-ment efficiency rates and replacement intervals.This methodology uses predictive technologies.

E. Equipment standardization=simplification: A phy-sical assets strategy is developed to include focuson simplifying and standardizing equipmentthroughout a plant. Successful implementation ofthe strategy reduces training and repair costs. Thisis particularly important in the ongoing ‘‘campaign’’approach of the typical batch pharmaceutical plant.

F. Reliability reporting: Special training is offered tomaintenance supervisors and workers to show themhow to measure and track reliability for equipmentin their areas. The focus is on building reliability

Figure 3 Typical pharmaceutical plant.

332 Oliverson

Page 348: Active Pharmaceutical Ingredients

indices for key equipment such as pumps, compres-sors, and motors. Assistance is provided on helpingpeople establish overall equipment effectiveness(OEE) and mean time between repair (MTBR) mea-sures for their areas.

G. Concurrent engineering: New engineering techni-ques are implemented to ensure that engineeringprojects are not developed in isolation. Cross-functional teams (engineering, production, mainte-nance, etc.) are involved in the design, installation,and testing of new equipment to ensure that relia-bility, maintainability, standardization, perfor-mance, and cost specifications for new equipmentare met. It is also very important to coordinateengineering efforts at the corporate, plant, andarea levels. During a shutdown in a pharmaceuti-cal plant, I observed three different maintenancecrews attempting to accomplish three distinctly dif-ferent modifications to the same unit based oninstructions from three separate engineering func-tions within the company.

H. Bottleneck study: Continuous flow manufacturing(CFM) techniques are used to identify manufacturingbottlenecks, especially those caused by inadequatereliability or maintenance practices. By prioritizingthe most critical bottlenecks, appropriate resourcescan be applied to maximize production throughput.

VIII. RISK MANAGEMENT

The best pharmaceutical plants focus on excellence in mainte-nance and reliability as a means of achieving manufacturingexcellence. This also results in compliance with environmentaland safety regulations and preserves a plant’s capital invest-ment. Techniques that minimize risks and surprises are:

A. Environmental=safety integration: This is a metho-dology ensuring that maintenance practices are

Plant Maintenance 333

Page 349: Active Pharmaceutical Ingredients

oriented to satisfying all environmental=safety regu-lations and requirements. The process involvesoperations, maintenance, engineering, safety, andenvironmental personnel in the design of best prac-tices. Maintenance personnel take a greater role ineducating operators on how to start up, operate, shut-down, and maintain their equipment more safely. Theprocess addresses EPA and OSHA (PSM) regulationsand requirements.

B. Life expectation study: This technical analysis iden-tifies the expected life cycle of critical equipment.Equipment life expectations are developed based onequipment histories, databases, and the manufac-turers’ information.

C. Life extension engineering: Production, maintenance,and engineering groups work together to devise meth-ods of extending equipment life. Best practices formaintaining equipment reliability and life are devel-oped, implemented, and measured. This methodologyis a natural outflow of the life expectation studies.

IX. SUMMARY

Excellence in the maintenance and reliability arenas is bestachieved by pursuing a logical, methodical approach, suchas we have outlined in this chapter. The pharmaceuticalindustry must move away from views such as ‘‘maintenanceis a necessary evil,’’ or ‘‘maintenance is an art,’’ to a reasonedempirical approach. The industry cannot afford to squanderits heavy investment in research and development by neglect-ing the maintenance and reliability function.

ACKNOWLEDGMENTS

I wish to acknowledge the contributions of my HSBRT collea-gues, Andy Ginder, Keith Burres, Kathy Sorensen, andShannan Porter in the development of this chapter.

334 Oliverson

Page 350: Active Pharmaceutical Ingredients

Index

Action letter, 172Active pharmaceutical

ingredients, 235Analytical methods, 213Approved for construction

version, 99Atom economy, 40Audit, 318

Basis of design, 142Best of breed approach, 317Biobatch, 22Biologicals, 13Biosynthesis process, 32Boil-outs, 223closed system, 226open system, 227

Bulk pharmaceuticalchemical, 205

Cephalosporins, 252Certificate of analysis, 241, 308Certificate of suitability, 199CGMP-compliant facilities, 237Change control, 210equipment change control, 210process change control, 210

Charge variance, 298Check-test-decide responsibility,

335Chemical reactions, 211Chemical entities, 13Chemical process, 15Chemists, 286industrial, 286medicinal, 27process, 27

Chemistry, 10bulk drug, 13

Class 100 conditions, 147Class number, 292class 100, 293class 10,000, 293

Classes, 292Classical fermentation, 247Clean room, 295Compressed gases, 251Consolidation stage, 24Contaminated product, 295distribution requirements

planning, 315extraneous contamination, 295uniform contamination, 295

Contamination, 4

335

Page 351: Active Pharmaceutical Ingredients

Continuous flowmanufacturing, 333

Current good manufacturingtechniques, 317

Design, basis of, 142Designing quality into

the process, 242Development stage, 23Distribution, 315Documentation, investigational,

170

Endotoxins, 252Environmental=safety integration,

334Equipment calibration, 209Equipment change control, 210Equipment qualification, 209

Factory acceptance test, 160Failure mode study, 331Failure modes, 331Fast tracking, 155initial conceptual estimate, 157

Fine chemical manufacturing, 1

Green chemistry, 30Good manufacturing practices

(GMP), 295

Hazard, 74High potency facilities, 136Hydroclones, 46

ICH Q7A, 144In-process controls, 262In-process testing, 185, 256Industrial chemists, 286aseptic processing, 292stagger shift operation, 289

[Industrial chemists]terminal sterilization, 292three-shift rotating

arrangement, 28912-hr shift cycle, 290

Installation qualification, 163Investigational documentation, 170ISO 9000, 318

Kilo lab, 25medicinal chemists, 27process chemists, 27

Launch platform plant, 114almost virtual companies, 117virtual drug companies, 117

Life cycle concept, 206concurrent approaches, 208prospective approaches, 208

Life expectation study, 334Life extension engineering, 334

Marketing application, 180Material, 13

On-condition system, 329Onsite waste treatment facility, 303Original investigational application,

169Outsourcing, 307

Performance measure, 325supportive key performance

measures, 328Performance qualification, 210Pharmaceutical industry, 2Pharmaceutical ingredient, active,

235Pharmaceulical manufacturing, 1Pharmaceutical process, 15Phase purity, 124

336 Index

Page 352: Active Pharmaceutical Ingredients

Preapproval inspection, 23Preliminary scope, 142Preparative stage, 23Prescription Drug User Fee Act, 193centralized procedure, 194mutual recognition

procedure, 194Prior approval supplement, 198BACPAC I guidance, 198BACPAC II, 198

Procedure, 229Process, 23, 205Process and instrumentation

diagram, 99Process change control, 210Process design, 96process design team, 99process development team, 99

Process documentation, 210Process validation, 210Product gross margin, 297fixed cost, 297labor rate variance, 298labor use variance, 298spending variance, 299variable cost, 297volume variance, 298yield variance, 298

Production planning, 306Proof of knowledge of control, 243Purchasing, 308Pure room, 215

Quality, overall summary, 191

Rapporteur, 194Reefers, 316Reengineering, 314Reference member state, 194november 1999 guidance, 197Type I variations, 196Type II variation, 197Type IA, 197

Type IB, [Reference memberstate]

197Reliability-balanced scorecard, 325Reliability-centered maintenance,

331Reverse auctions, 310negotiation brief, 310

Risk, 74Route, 23

Scale-up, 42Scheme, 23Screening programs, 5Semisynthesis, 14chemical synthesis step, 15

Specifications, 309Staggared shafts, 289Sterilization, terminal, 292Sterilization-in-place procedure,

229Strain mutation, 33

Techniques, 333Technology transfer, 106Technology transfer stage, 24Thalidomide,Transportation, 315

Unidirectional flow, 147Unit operations, 17, 211, 212

Validation, 205Virtual companies, 117

Walk down, 163Warehousing, 136Workshifts, staggered, 289

Index 337

Page 353: Active Pharmaceutical Ingredients
Page 354: Active Pharmaceutical Ingredients