82
Reprinted from PHARMACEUTICAL ENGINEERING® The Official Journal of ISPE March/April 2003, Vol. 23 No. 2

Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

  • Upload
    others

  • View
    16

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Reprinted from

PHARMACEUTICAL ENGINEERING®

The Official Journal of ISPE

March/April 2003, Vol. 23 No. 2

Page 2: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

2nd Annual ISPE Japan Affiliate ConferenceJune 12-13, 2003

Tokyo, Japan

Join ISPE at the 2nd Annual Japan Affiliate Conference June 12-13 inTokyo, Japan. The Conference will discuss important issues and

challenges facing the industry and will feature key seminars based on themost pressing issues in today’s Japanese Life Science Industry. Confer-ence delegates will benefit from discussions by speakers from around theworld on current trends in regulatory issues, the ISPE Baseline® GuideSeries, GAMP 4, Barrier Isolation, and Technology Transfer.

ISPE Japan Affiliate LeadershipPresidentTomiyasu Hirachi, Shionogi & Co., Ltd.e-mail: [email protected]

Director, Head of Secretariat, and Chairmanof Organization CommitteeShinichi Osada, JGC Corporatione-mail: [email protected]

Director, Finance Officer and Chairman ofPublications CommitteeHiroshi Kyogoku, Chiyoda Corporatione-mail: [email protected]

Director, Special AdvisorYoshizo Fukuda, Japan NUS Co., Ltd.e-mail: [email protected]

Director and Chairman of RegulatoryCommitteeKeiji Yamamoto, Chiba University, PostGraduate School of Pharmacye-mail: [email protected]

Director, Chairman of CollaborationCommitteeKoji Aichi, Taisho Pharmaceutical Co., Ltd.e-mail: [email protected]

Director and Chairman of Discussion ForumCommitteeTakeshi Takashima, Powrex Corporatione-mail: [email protected]

Director and Chairman of Education andTraining CommitteeTakeshi Harima, Pfizer Pharmaceuticals, Inc.e-mail: [email protected]

Director and Chairman of Winter ConferenceCommitteeYoshiaki Kawashima, Gifu University, Schoolof Pharmacye-mail: [email protected]

Director and Vice Chairman of WinterConference CommitteeYasuo Miyake, Chiyoda Corporationemail: oripharm-jpn@k7

Page 3: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

Dear ISPE Member:

The origins of Japan’s pharmaceutical history go backover one thousand years with its notable period ofgrowth occurring in the Edo Period (1603-1868).Developing in several distinct regions of the country,pharmaceuticals were soon to become a leading indus-try.

Now, with its graying population, there is an increasingneed in Japan for new pharmaceutical products. Thiswill further drive the requirement for development ofquality R&D as well as production sites in Japan.

As local manufacturers set their sights on supplyingdomestic and export markets, and as the major globalfirms invest in a long-term presence in Japan, regula-tory harmonization and compliance will be of criticalimportance.

Meanwhile, the matter of optimal sharing of informa-tion is being addressed. As an example, seminarspresented by visiting Life Science professionals hostedby the Japan Affiliate are simultaneously interpreted,while the translation of ISPE publications into theJapanese language is being energetically promoted.

We trust that our Japan Country Profile will be ofinterest to you and contribute to an understanding ofthe background against which the Affiliate very re-cently emerged.

Yours truly,

Tomiyasu Hirachi

Tomiyasu HirachiChairman, ISPE Japan Affiliate

This new feature inPharmaceuticalEngineering isdesigned so thatyou can tear it out,three hole drill(if desired), andkeep it with otherCountry Profiles asthey are published.

Look for theCountry Profile onthe Nordics in theMay/June issue ofPharmaceuticalEngineering.

Photographs courtesy ofTakayuki Hayashida,ISPE Member.

Page 4: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Japan - the World’s Second LargestPharmaceutical Market

Figure 1. Japan’s pharmaceutical market.

Figure 2. Share of world pharmaceutical market (2001).

For more than 10 years,Japan has been one of theworld’s largest pharma-ceutical markets with

sales totaling about $56 billion infiscal year 2000. Figure 1 showsthe changes in sales of pharma-ceuticals and ethical drugs from1990 to 2000; the growth rate forthis 10-year period was 11.2%.

Moreover, the size of the marketfor ethical drugs alone was about$49 billion, representing some13% of the world total market(Figure 2) and putting Japan insecond place after the UnitedStates. For comparison, pharma-ceutical sales in Japan compriseabout 54% of sales in all of Eu-rope.

Table A shows the growth fore-cast by IMS Health—the Japa-nese market is expected to seegrowth of about 2.3% and thenation is expected to remain theworld’s second largest marketoverall with some 15.1% of thetotal world market share.

Recognizing this important sta-tus and with the need for furtherstimulus in mind, the Japanesegovernment has published its vi-sion of the future of the Japanesepharmaceutical manufacturingindustry, outlining three majorthemes:

• Create a good environment forthe development of effectivenew drugs. In particular, aimto establish systems for devel-oping, testing, and licensingnew drugs based on genomeR&D to bring better ethical

products to market morequickly than at present.

• Plan to revise the Pharmaceu-ticals Affairs Law.

• Plan for growth in the genericand OTC drug markets.

Japan’s rapidly aging society isexpected to see large increases inmedical expenses. Also, planned

growth of Japan’s pharma-ceutical industry, based on thegovernment’s future vision, sug-gests that the industry has sub-stantial potential.

Status of Japan’sPharmaceutical

Manufacturing IndustryJapan’s pharmaceutical manufac-turing industry is characterizedby a very large number of small

Page 5: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

manufacturers. In addition, therehas been a recent increase in for-eign investment after the market-opening measures. Moreover, com-panies in other business fields areentering pharmaceuticals by de-veloping their own technologies.Although the market is large, theframework of the National HealthInsurance (NHI) system makesfuture expansion likely to see morecompetitors scrambling for a shareof the pie. New drugs are beingdeveloped to compete with theoverseas pharmaceutical giantsand Mergers and Acquisitions(M&As) are being examined as ameans of surviving in an increas-ingly competitive market place,but positive activity seems moreprobable in the future.

Size and Structure ofthe Pharmaceutical

Manufacturing IndustryIn 1997, Japan’s pharma-ceutical manufacturers pro-

duced about $51 billion ofproducts, accounting for about1.02% of Gross Domestic Prod-uct (GDP) and 1.9% of all manu-factured products. The propor-tion rises to 3.4% of all manu-

factured products in terms ofvalue added through payments ofsalaries to employees and taxesto government, demonstrating the Figure 3. Contribution of pharmaceutical to GDP (1997).

Total Sales Forecast Sales Forecast Annual Market2000 2005 Growth Share

(US$ Billion) (US$ Billion) 2000-05 (%) (%)

USA 150 263 11.8 60.5

Japan 58 66 2.3 15.1

Germany 17 24 7.5 5.6

France 16 22 6.0 5.0

UK 11 16 8.3 3.7

Italy 11 16 8.2 3.7

Canada 6 10 10.7 2.3

Spain 6 10 9.9 2.3

Australia 3 5 9.3 1.1

Belgium 2 3 5.6 0.7

Total 280 435 9.1 100.0

Table A. Growth forecast for top 10 world markets (2000).

large role of the industry inJapan’s economy.

Employee numbers increased un-til 1985, but have stabilized ataround 2,000,000 since then,comprising about 0.3% of Japan’stotal labor force and 1.4% of themanufacturing industry. Theworkforce is relatively small incomparison to the impact of theproduction value on the Japa-nese economy.

Looking at the market shares ofthe world’s top 31 companies withtotal sales exceeding $20 billion,12 US companies account for morethan 50% of total sales, followedby two UK companies with 15.4%,

and two companies each in Swit-zerland and France with 9% and8%, respectively. The top sevenJapanese manufacturers had an8.4% share—these seven busi-nesses are all of similar size andjostle for position in the crowdeddomestic marketplace. However,there are big differences betweenthe top seven and the other lower-ranked companies.

About 80% of some 1400 pharma-ceutical manufacturers in Japanare capitalized at under $2.5 mil-lion and 60% are medium or smallcompanies with total annual salesof less than $2.5 million. In addi-tion, about half have fewer than50 employees, 22% have fewerthan 10 employees, and only 8%have more than 1000 employees.On the other hand, the top 50companies account for about 87%of annual pharmaceutical sales.

The 18 foreign-capitalized com-panies have about a 25% share ofsales; the number of these com-panies and the size of their mar-ket share are both expected toincrease in the future.

Special Featuresof Japan

A unique feature of the Japanesepharmaceutical business is thetraditional practice of salesmenvisiting private households once

Page 6: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 4. Market share of world’s top 31 pharmaceutical companies.

Value of Shipments Value Added(US$ million) (US$ million)

Pharmaceuticals 51,128 33,809

Soft Drinks 18,940 8,311

Oil Refining 67,528 4,949

Steel Manufacturing 38,256 16,633

Household Appliances 34,488 13,712

Computers 83,861 18,128

ICs 60,669 20,581

Automobiles 177,491 43,306

(Source)MITI 1998 Table of Industry Statistics

Table C. Comparison of pharmaceutical and other industries.

a year to leave a medicine box andcharging only for medicines thathave been used in the previousyear when replacing them at thenext annual visit. This unusualsales technique is a remnant ofbusiness practices from themiddle Edo period 200 or 300years ago, and is typically found

in Toyama Prefecture. Businessesmaking and selling OTC drugscomprise the largest number ofJapanese pharmaceutical compa-nies, but account for less than12% of production value.

In Japan, the government controlsthe prices of ethical drugs. Unlike

the United States and Germanywhere businesses can choose high-or low-price strategies, Japanesemanufacturers do not have free-dom to set the prices of controlledproducts. In addition, ethical drugprices are continually forced down.This is clearly different from theUnited Sates and Germanywhere prices for the same prod-ucts either increase or remainstable. Furthermore, the impact ofgeneric drugs is relatively limitedin Japan unlike the United Statesand Germany where the arrival ofgenerics on the market has a largeimpact on branded drugs.

Meeting FutureChallenges

The ballooning development costsfor new ethical drugs cannot berecovered from domestic salesalone, and a major issue iswhether or not to enter overseasmarkets. Success or failure of thebusiness is clearly dependent onthe sales growth of new drugs foroverseas markets. On the otherhand, foreign-capitalized busi-nesses are becoming more activein the domestic market and therewill undoubtedly be more depen-dence on foreign capital in thefuture. This process is leadingthe pharmaceutical industry to-ward adoption of various globalunified rules and standards,meaning that there will be noroom in the future for just domes-tically focused management poli-cies. Some are of the opinion thatit will be difficult even for largemanufacturers to follow an inde-pendent path considering the con-ditions of the global market, butit might be possible to surviveindependently by choosing theright strategy. One possibility isreorganization, and many Japa-nese pharmaceutical manufactur-ers are taking steps in this direc-tion. This competitive weeding-out is likely to lead to better in-dustrial productivity and an effi-cient business structure.

Amount Proportion of Proportion(US$ Billion) GDP Industry

Market Size (1997) 61.9 1.5% -----

Value (1997) 51.2 1.2% 1.9%

Value Added Pharmaceuticals (1997) 33.8 0.8% 3.4%

(Sources)OECD Health Data“Value”: Ministry of Health, Labour and Welfare ‘Pharmaceutical Industry Production Statistics’“Value Added Pharmaceuticals”: MITI Table of Industry Statistics

Table B. Role of pharmaceutical industry in Japanese economy.

Page 7: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

Figure 5. Structure of Japan’s pharmaceutical industry.

As a result, we can expect to seethe appearance of manufactur-ing giants who can bear the bur-dens of massive R&D costs byoffering revolutionary new drugsto the world’s markets. On theother hand, we will probably seean increase in the growth of ge-neric drug companies respondingto the increasing need for low-cost, good-quality ethical drugs.

Moreover, since R&D results canbe shared in the pharmaceuticalindustry, relatively small-scalebusinesses also have an opportu-nity to achieve major results.

There are also many in-stances where medium-level

businesses can use these op-portunities to achieve growth intheir specialist fields.

PharmaceuticalDevelopment in Japan

Many New Ethical DrugsDiscovered in JapanDevelopment of the Japanesepharmaceutical industry started

in earnest after the postwar re-covery. Japanese society changedgreatly with economic growth andthe NHI system was introducedin 1961 creating a surge in de-mand for pharmaceuticals. In linewith these changes, there was aneed for new and more effectivedrugs. As a result, Japanese phar-

maceutical manufacturers fo-cused their attention in the shortterm on introducing and market-ing products from the US andEurope while establishing theirown R&D systems and improvingtheir technical abilities in newdrug development.

Page 8: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

8 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 7. Pharmaceutical shipments by foreign-capitalized companies.

Figure 6. Japan’s pharmaceutical manufacturers based on sales.

Figure 8 shows the nationalshares of new international ethi-cal drugs in the last 20 years.

Table D. Pharmaceutical manufacturers by number of employees.

Japan (Unit:%)

Fiscal Year 10 employees 11~50 51~100 101~300 301~1,000 1,001~3,000 Over 3,000 Totalor less employees employees employees employees employees employees

1980 24.9 40.2 9.7 12.0 7.0 3.5 2.7 100.0

1985 27.4 33.4 12.0 13.6 7.2 3.6 2.8 100.0

1990 29.4 32.4 10.7 13.7 6.7 5.0 2.2 100.0

1995 25.6 30.3 11.2 16.4 8.0 5.1 3.4 100.0

1999 22.3 32.7 11.5 16.8 8.7 4.8 3.2 100.0

2000 JPMA ----- ----- 1.2 4.8 27.4 41.7 25.0 100.0(84 Members)

(Sources)“Pharmaceutical Survey” by Ministry of Health and Welfare. UP to FY 1998.“Pharmaceutical and Medical Device Industry Survey” by Ministry of Health, Labour and Welfare FY 1999.

Table E lists new ethical drugsdiscovered in Japan.

This data indicates that Japan’spharmaceutical industry has dis-covered a large number of newethical drugs, and that, clearly,the ability to develop originaldrugs has increased greatly sincethe 1980s.

For the present and in the nearfuture, we can expect attention tobe focused on the development ofnew drugs based on data from thehuman genome project. R&D ac-tivities in new ethical drugs arebeing revolutionized by integra-tion and improvement of a widerange of leading-edge technolo-gies and knowledge including ge-nome research, protein research,bioinformatics fusing IT, and bio-technology—as well as optimiz-ing and screening of compounds,technologies, etc. Development ofnew ethical drugs requires in-creasingly large investments,driving worldwide M&As leadingto the birth of pharmaceuticalgiants with the requisite finan-cial and human resources.

Japan has lagged slightly behindthe leaders in genome analysis—investment by the Japanese phar-maceutical industry comparesunfavorably with the major in-ternational pharmaceutical com-panies. If this trend continues,there are worries that the Japa-nese pharmaceutical industry will

Page 9: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 9©Copyright ISPE 2003

be weakened in the future. TheJapanese government has there-fore established a national projectbased on further improvementsin drug safety to create a favor-able environment for developingoriginal ethical drugs for interna-tional markets and to strengthenthe competitive power of thenation’s pharmaceutical manu-facturers. Clearly, the 21st cen-tury will be the age of IT andbiotechnology; Japan’s pharma-ceutical industry is being nur-tured to become one of its leadingindustries and is increasinglyimportant in the overall develop-ment of the Japanese economy.

R&D Investment- More than 12% of SalesDevelopment of useful new ethi-cal drugs is a key factor in growthof the pharmaceutical industry.

Japan’s principal pharmaceuti-cal manufacturers invest almost13% of total sales revenues inR&D and there are indicationsthat the ratio is rising.

Figure 9 compares the change inR&D investment between Japanand the US since 1990. The ratioto sales has remained at a rela-tively high level.

Figure 8. Share of new international ethical drugs.

Page 10: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

10 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Bringing aNew Ethical Drug

to the MarketTakes More

than a DecadeData from the Japan Pharmaceu-tical Manufacturers Association(JPMA) shows that 15 to 17years are required from thestart of R&D to final approval,and bringing the new drug to themarket. Furthermore, the chanceof a candidate compound obtain-ing approval is one in 11,300 or0.009%. As shown in Figure 10,the success rate from the start ofpre-clinical testing until a productcomes on the market rises to 0.13%with a minimum R&D investment

Table E. New ethical drugs discovered in Japan.

Company Nonproprietary Name Indication Launch (Year) Sale (Country)

Eisai Rabeprazole Na Peptic ulcer 1998 3Donepezil hydrochloride Alzheimer 1997 36

Ono Alprostadil Obstructed peripheral artery 1985 17Gemeprost Prostglandines 1984 15

Sankyo Troglitazone Diabetes 1997 7Cefpodoxime Infection 1991 58Pravastatin Hyperlipemia 1990 72

Shionogi Ceftibuten Infection 1992 46

Takeda Candesartan Hypertension 1997 >10Lansoprazole Peptic ulcer 1991 >80Leuprorelin Prostate cancer 1989 >60Cefotiam Infection 1981 >10Citicoline Disturbance of consciousness 1970 >20

Tanabe Imidapril hydrochloride Hypertension 1995 5Diltiazem hydrochloride Hypertension 1977 >100

Daiichi Lavoflxacin Infection 1997 25Ofloxacin Infection 1985 140

Tranexamic acid Allergy 1965 100

Dainippon Sparfloxacin Infection 1994 18Enoxacin Infection 1987 17

Chugai Lenograstim Leukocytopenia 1993 55Nicorandil Angina pectoris 1993 11Sucralfate Peptic ulcer 1971 <100

Fujisawa Tacrolimus Graft rejection 1993 26Cefdinir Infection 1991 5

Nilvadipipine Hypertension 1989 5Cefixime Infection 1987 77

Ceftizoxime Infection 1982 43Zotepine Schizophrenia 1982 3Cefazolin Infection 1971 60

Yamanouchi Famotidine Peptic ulcer 1986 112Nicardipine hydrochloride Hypertension 1984 56

Josamycin Infection 1980 67Cefotetan sodium Infection 1984 11

Tamulosin hydrochloride Dysuria 1995 40Formoterol fumarate Bronchial asthma 1988 53

Page 11: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 11©Copyright ISPE 2003

Figure 9. R&D investment in Japan and US.

of $220 to 300 million and a timeinvestment of 11 to 12 years.

Hollowing-out ofClinical Trials?

Clinical trials are an indispens-able stage in the development ofnew ethical drugs and the costs ofclinical trials comprise about 40%of the total R&D investment inany new drug.

Looking at the situation in Ja-pan, although R&D investmentis increasing, the number of ap-

plications to conduct clini-cal trials in Japan is de-

creasing. This is due to theimpact of trial-related changes

to regulations, such as new drugpricing evaluation, new GoodClinical Practice (GCP) en-forcement, and expanded ac-ceptance of data from over-

seas clinical trials—Japanesepharmaceutical manufacturersare focusing on development of

new international ethical drugsto come on the market first in theUnited States and Europe andare changing the trial environ-ment by placing greater priorityon trials in these countries. Inother words, there is a hollowing-out of clinical trials in Japan.

This hollowing-out of clinical tri-als is having a certain level ofimpact on Japan’s publichealthcare and the internationalcompetitiveness of its pharma-ceutical industry. The govern-ment has therefore includedchanges to the clinical-trial envi-

Figure 10. Success rate and time required to develop branded drugs.

Page 12: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

12 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 11. Clinical trial applications.

Figure 12. Ethical drugs under development in Japan.

ronment as one item in the na-tional action plan.

Foreign ProductsMake Up 44%

of Japan’sPharmaceutical Market

Currently, about 44% of pharma-ceuticals sold in Japan are for-eign products—no other marketin Japan is as open as this.

The trade balance of pharmaceu-ticals in Japan is showing aslightly decreasing trend, butthere is still an excess of imports.

Conversely, the trade in pharma-ceutical-related technology hasbeen overwhelmingly export ledsince the mid-1990s, demonstrat-

ing the high technological levelof Japan’s pharmaceutical indus-try.

Hopes and Fears -Focusing on Japan’s

PharmaceuticalManufacturing

Environment andIndustry

In the short term, Japan’s phar-maceutical industry is endeavor-ing to ensure its ability to de-velop new ethical drugs. Devel-oping new effective drugs thatcontribute to healthcare whilesimultaneously having an impacton Japan’s economic growth pre-sents a number of issues. Againstthis background, the governmentcanvassed opinions from various

medical-related bodies, manufac-turers’ associations and learnedpersons. In August 2002, it fi-nally incorporated a Vision forthe Pharmaceutical Industry in anational action plan aimed atdeveloping a future image for theindustry and promotinggrowth.

The items related to new drugdevelopment are listed below.

1. Strengthen systems for sup-porting fundamental researchwith the intention of discover-ing new ethical drugs and ac-quiring excellent technologies.

2. Strengthen systems for trans-ferring results and technolo-gies from national research or-ganizations to private compa-nies and for assisting long-term cooperation between in-dustry, government, andacademia.

3. Make changes to regulatoryenvironment controlling clini-cal trials.

4. Create a responsive body forapproving new ethical drugs.

5. Establish a NHI drug pricesystem that encourages inno-vation.

Attention should be given to thefuture Japanese pharmaceuticalmanufacturing environment andindustry.

Japan’s Medical andPublic Welfare SystemsOverviewIf Japan’s medical and publicwelfare system could be summedup in one phrase, it would beNHI. This uniquely Japanese sys-tem was set up in 1961; it is apublic medical health insurancesystem for ensuring that every-body in Japan has fair and equal

Page 13: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 13©Copyright ISPE 2003

Figure 13. Pharmaceuticals trade balance.

Figure 14. Pharmaceutical technology exchange.

Figure 15. Average Japanese life expectancy.

access to medical treatment. Anadvantage of the system is notthat everybody who has joinedthe system receives the very bestmedical care, but that anybodycan access average care at anytime. Japanese now have the long-est average life expectancy in theworld, possibly reflecting the suc-cess of the NHI system. On theother hand, Japan’s rapidly ag-ing society means that the nationfaces rising medical costs andworsening NHI financial re-sources that will require drasticsolutions.

National Medical CostsThe facts of healthcare in Japancan be examined based on datafrom the NHI system.

As shown in Figure 16, due to therising life expectancy and im-provements in medical technol-ogy, annual national medical costsare increasing steadily and havealready passed $250 billion—anearly 50% increase in 10 years.A unique characteristic of the datais that about 35% of the totalcosts is medical care for peopleaged 70 or older. The Ministry ofHealth, Labor, and Welfare(MHLW) estimates that the pro-portion of the elderly populationwill increase from 11.8% atpresent to 21.7% by 2025 whenthe nation’s medical costs willexceed $670 billion—56% of thisamount will be for geriatric care.In these circumstances, there willbe tremendous pressure to forcemedical costs down, meaning a

reversal in the proportion ofcosts for pharmaceuticals.

There has already been a 10%drop in the proportion over thelast 10 years - Figure 16. Incomparison to the runaway to-tal medical costs, the total forpharmaceuticals has re-

mained broadly similar for thesame 10-year period.

Page 14: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

14 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 17. Pharmaceutical price Indices in Japan, US, and Germany.

Price ofPharmaceuticals

As described above, the marketconditions for ethical drugs areextremely severe. One reason isthat unlike the US and Germany,Japanese pharmaceutical manu-facturers are limited in their powerto set prices freely. In Japan, thegovernment sets the prices of new

Figure 16. National medical costs and proportion of pharmaceuticals.

drugs coming on the market basedon the NHI drug price system whilethe actual price set by the manu-facturers is always higher thanthe government price.

The data in Figure 17 demon-strate this situation very well—only Japan has seen a drop in thepharmaceutical price index.

Reforming MedicalSystems

Japan is an aging society andstatistics for 2000 show that Japa-nese women are only having 1.36children on average. In addition,figures published in 1996 for the29 Organization for EconomicCooperation and Develop-ment (OECD) nations showthat in Japan, the average hos-pital stay is 33.5 days. These cir-cumstances are putting a verysevere strain on the financial re-sources of the health insuranceassociations supporting the NHIsystem. Although the privatehealth insurance organizationshave been said to have a rela-tively good surplus of resources,some 78% actually had businessdeficits in fiscal 2001.

In these circumstances, the gov-ernment is starting to make con-crete proposals for drastic reformsof the Japanese healthcare sys-tem. Such reforms are bound tohave a major impact on the marketfor ethical drugs in Japan and the

Page 15: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 15©Copyright ISPE 2003

Figure 18. Healthcare system reforms.

business plans of Japanese phar-maceutical manufacturers.

ConclusionThe quality of pharmaceuticalsproduced by Japanese manufac-turing plants is second to noneand a great many resources areinvested in quality assurance. Onthe other hand, the pharmaceuti-cal industry is rapidly becomingglobalized and we have enteredthe age when internationally rec-ognized quality standards are re-quired.

Furthermore, drastic reforms willbe required to sustain healthcaresystems like Japan’s NHI systembased on medical insurance.

In this type of business environ-ment, Japanese pharmaceuticalmanufacturers are gradually ex-panding their international busi-ness presence while doing theirbest to offer patients in Japanand around the world even saferand more effective new drugs inthe shortest possible developmenttimeframes.

Page 16: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Country Profile - Japan

16 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Representative Japanese OrganizationsActive in the Pharmaceutical Field

JPMAJapan Pharmaceutical ManufacturersAssociation

The Japan Pharmaceutical Manufacturers Associa-tion is a voluntary organization of research-basedpharmaceutical manufacturers that contribute tosociety by developing new pharmaceuticals.

As of October 2002, the JPMA had 80 members(including 22 foreign affiliates) and 11 committees.The JPMA works in close cooperation with the Inter-national Federation of Pharmaceutical Manufactur-ers Associations (IFPMA).

http://www.jpma.or.jp

JSPMEJapan Society of PharmaceuticalMachinery and Engineering

The Japan Society of Pharmaceutical Machinery andEngineering is a not-for-profit volunteer societyfounded in 1991 in order to advance pharmaceuticaltechnology through exchange of knowledge and expe-rience in a wide range of industries that are devotedto pharmaceutical production.

Members are scientists from academia, govern-ment, and companies involved in pharmaceuticals,machinery, construction, electronics, and computerswho are dedicated to obtaining the highest level ofquality and efficiency.

Merino Bld. 5F 1-28Kandasuda-cho Chiyoda-kuTokyo 101-0041, Japan

Tel: +81-3-3252-3048Fax: +81-3-3252-3049

E-mail:[email protected]

http://www.seikiken.or.jp

MHLWMinistry of Health, Labour, and Welfare

1-2-2 Kasumigaseki Chiyoda-kuTokyo 100-8916 Japan

Tel: +81-3-5253-1111

E-mail: [email protected]

http://www.mhlw.go.jp

OPSR/KikoThe Organization for PharmaceuticalSafety and Research

Establishment: October 15, 1979

Japanese semi-governmental organization autho-rized by the Minister of Health, Labour, and Welfare(MHLW).

Shin-Kasumigaseki Building, 9th Floor3-3-2 Kasumigaseki, Chiyoda-kuTokyo 100-0013 Japan

http://www.kiko.go.jp

Page 17: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

ISPE Baseline® Pharmaceutical Engineering Guide ForNew and Renovated Facilities Volume 6:Biopharmaceuticals - ExecutiveSummary

The followingExecutiveSummaryrepresents thedraft of the ISPEBiopharmaceuticalsBaseline® Guidethat has beenavailable to FDAand ISPEmembers forcomment, withcomments due toISPE by March2003.

It is likely thatparts of thissummary willchange to reflectthese comments,and therefore thesummary shouldbe consideredonly a generalindicator of thetopics covered inthe Baseline®Guide.

Table of Contents1 Introduction1.1 Background1.2 Scope of The Guide1.3 Key Concepts of the Guide1.4 Using the Guide2 The Regulatory Basis for Facility Re-

quirements2.1 Executive Summary2.2 Scope2.3 Definitions2.4 Regulatory Considerations2.5 General Concepts2.6 Bibliography2.7 Significant Regulatory Documents3 Manufacturing Operations and Activi-

ties

3.1 Executive Summary3.2 Open Versus Closed Systems3.3 Pyrogen-Controlled Processing3.4 Considerations For Multi-Product Op-

erations3.5 Viral Clearance3.6 Stage of Product Development3.7 Operational Upset3.8 Operability and Maintainability3.9 Cleaning and Housekeeping Consider-

ations4 Chapter 4: Process And Equipment4.1 Executive Summary4.2 Typical Biopharmaceutical Processes4.3 Critical Process Parameters4.4 General Considerations for Equipment

Design

Figure 1. Proposedstructure of the finalISPE Baseline® Guide forBiopharmaceuticals.

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 18: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

4.5 Specific Equipment Design Con-siderations

4.6 Summary5 Process Support and Utilities5.1 Executive Summary5.2 Regulatory Issues5.3 System Impact Descriptions5.4 System Layout and Routing5.5 Specific Service Considerations6 Facility6.1 Executive Summary6.2 Process Considerations6.3 Operational Considerations6.4 Facility Layout Considerations6.5 Operational Support6.6 Area Environment6.7 Architecture And Finishes6.8 Discretionary Considerations7 Process Controls7.1 Executive Summary7.2 Biopharmaceutical Automation

Issues7.3 Level of Automation7.4 Biopharmaceutical Unit Opera-

tions7.5 Control Systems Maintenance7.6 Validation of Biopharmaceutical

Automation Systems8 Commissioning and Qualifica-

tion8.1 Executive Summary8.2 Impact Assessment8.3 Qualification9 Glossary10 Appendix - European Aspects10.1 Introduction (General)10.2 Water Quality (Ref: Chapter 4

And 5)10.3 Bio-Containment and Environ-

mental Protection (See Chapter 5)10.4 Environmental Impact Issues10.5 Chromatography Skid Sharing10.6 Qualification and Validation

(Ref: Chapter 8)

1. Introduction1.1 BackgroundThe design, construction, commission-ing, and qualification of biophar-maceutical facilities will challengemanufacturers, engineering profes-sionals, and equipment suppliers.These facilities must not only meetcGMP regulations, but must complywith local codes, laws, and regulations.

The current situation is one of con-fusion and sometimes little science:

• Solutions are applied out of context(one product’s approaches inappro-priately applied to a different typeof product)

• Product and process are not consid-ered in decisions. A common reason is“Company X did it, so we should, too.”

Capital concerns:• Capital funds may be limited, so

wise use of funds is important• The need to get quick facility ap-

proval at all costs has led to over-spending to remove potential snagsduring inspections

Money is not going toward productprotection as much as to “fluff:”• Money that could have been used

for protecting the product is divertedto features with no product impact:- Mirror finishes, “stainless steel”

facilities- Confusion regarding required

process water quality, often over-specified without economic or sci-entific justification

- Classified spaces (cleanrooms)where they are not needed, as forclosed processes

1.2 Scope of the GuideThis Guide may be used by industry forthe design, construction, commission-ing, and qualification of new and reno-vated biopharmaceutical facilities. Itis neither a standard nor a GMP, nor isit a detailed design guide. It is notintended to replace governing laws orregulations that apply to facilities ofthis type. The application of this docu-ment for new or existing facilities is atthe discretion of the facility owner oroperator. Approaches to meeting GMPprovided in this Guide need not beretroactively applied to currently li-censed facilities.

This Guide applies to large mol-ecule biotech products, cell-cultured,or fermented:• It does not apply to blood, vaccines,

etc. However, most concepts in thisGuide may be applied to these prod-ucts.

• It applies to biopharmaceutical Ac-tive Pharmaceutical Ingredient(API) products licensed by both theCenter for Biologics Evaluation andResearch (CBER) and Center for

Drug Evaluation and Research(CDER).

US GMPs:• Not much is specifically stated in the

US GMPs, but best practices arecovered here. It is ultimately theowner’s responsibility to justify deci-sions and approaches to regulators.

Other GMPs are covered in the Appen-dix. National Institute of Health (NIH)and other safety issues are mentionedin the Guide where they affect GMPs ordesign.

The audience for this Guide is pro-fessionals involved in the design, con-struction, validation, and operation oflicensed biopharmaceutical manufac-turing facilities.• The mission of the Baseline® Guides

is to help operating companies sat-isfy the GMPs and produce productin a manner that allows the manu-facturer to stay in business.

• This Guide is not a GMP, but in-stead it focuses on the use of re-sources to meet GMP. This Guide isbut one approach to satisfying theintent of the GMPs. Other methodsof protecting the product may existnow or evolve in the future. If anissue is not covered in this Guide, orif alternatives appear feasible, thereader is advised to discuss themwith the appropriate regulatoryagencies before significant finan-cial commitments are made.

• It is intended that this Guide will beused by regulators and quality con-trol personnel to understand the tech-nical issues regarding the facility orprocess. This Guide does not attemptto cover the basics of the engineeringsciences, nor does it attempt to coverbiopharmaceutical GMPs that do notaddress the facility or the manufac-turing process technology.

1.3 Key Concepts of the Guide1.3.1 Does the Process EqualProduct?There is a continuum of process andfacility approaches based on the prod-uct and processes used to make theproduct. The best engineering solutionmakes optimal use of people, materials,and capital while protecting the prod-uct. There is not one “right” or “perfect”

Page 19: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

way to design and operate the facility.However, the design of a facility has aprofound impact on process design andon how the facility is operated.

Due to limitations in analytical meth-odologies and only superficial under-standings of the relationships betweenprocess variables and final product qual-ity, biopharmaceutical processes havehistorically been viewed as “blackboxes.” Thus, there also has been aprevailing view that the “process equalsthe product.” This view has led to reluc-tance to alter biophar-maceutical pro-cesses, a reluctance that has been rein-forced by conservative regulatory ap-proaches. How could manufacturersassure the identity of the final productin the case of process variations? Howcould manufacturers assure the finalproduct with changes in scale or changesin the facilities of manufacture?

As the industry has developed abetter understanding of biophar-maceutical processes and as analyticalmethods have improved, we have de-veloped a better understanding of the“cause and effect” relationship betweenprocess variables and products. Thisevolution has caused a change in focusto those issues that are critical to theconsistent manufacture of high qualityproducts. Products and processes havebeen proven to be transportable be-tween facilities and can be operated ondifferent scales.

1.3.2 Process Design is Tiedto Facility DesignThis Guide covers the variables thatmost directly affect the process andfacility:• Open versus closed processing:

- Closed processing places moreemphasis on protecting the prod-uct INSIDE the process.

- Open processing places more em-phasis on the facility and itspeople.

• What works best for one product,facility, or process scale may notwork best for another product, facil-ity, or process scale.

• Features that work well in a singleproduct facility may be inadequatefor a multiple product facility.

• Chapter 3 discusses these issues as

well as viral clearance and clinicalmaterials manufacture.

Process controls:• Automation is not a GMP require-

ment, but if automation is used,then there are GMP implications.Chapter 7 provides more insightregarding automation.

For subjects generic to all pharmaceu-tical facilities, the reader is directed toother sources for more in-depth infor-mation.• Qualification basics are covered in

the ISPE Baseline® Guide for Com-missioning and Qualification.- Commission everything in accor-

dance with Good EngineeringPractice, but qualify only directimpact systems and critical com-ponents of those systems.

- Design Qualification or En-hanced Design Review will helpcomply with ICH Q7A.

- Qualification considerations spe-cific to Biopharmaceutical sys-tems are covered in Chapter 8with reference to topic-specificqualification activities in Chap-ters 3 through 7.

• Water and steam systems are cov-ered in the ISPE Baseline® Guidefor Water and Steam Systems.

The Guide user is encouraged to workwith the regulators to iron out “unique”issues before they become significantissues.

1.3.3 Controlled ProcessingThe product must be protected by con-trolling the process and often its sur-roundings. This requires knowledge ofthe product and process and protectionutilizing segregation and flow patterns.Chapter 3 discusses controlled process-ing in more detail.

1.3.3.1 Know the Product(and its Process)Intimate knowledge of the product, itscritical parameters, the processes in-volved, and processing parameters isessential. Evaluation of potential con-tamination routes is needed. Data thatdemonstrate control of the process andto justify processing decisions will bekey to a successful facility.

1.3.3.2 The Process CannotAdd ContaminationThe process’s contamination profilemust be known and the process con-trolled to specifications.• Process Water should reflect the

product purity profile.• Chapter 3 discusses recovery from

upsets and prevention of contami-nation in manufacturing operations.

1.3.3.3 Contamination ControlStrategyAs discussed in Chapter 3, bulkbiopharmaceutical manufacturing islow “bioburden” production. Aseptic-like processing steps or “sterile” pro-cessing operations utilizing sterilizedprocess equipment are usually oper-ated closed.

Chapter 3 also discusses housekeep-ing, cleaning, and fumigation. Chapter4 discusses equipment cleanability, andclosure.

1.3.4 Segregation and FlowSegregation protects the product fromcontamination in its surroundings (i.e.,from the facility and other products).Segregation may be accomplished viaprocedures, timing, or by physicalmeans. Flow patterns in the facilityinfluence segregation, especially if morethan one product is manufacturedthere. Chapter 6 provides more detailto help decision-making regarding seg-regation and flow.

Primary and secondary segregation:As discussed in Chapters 2, 3, and 6,protection of product may be accom-plished through primary and second-ary segregation.

Primary Segregation - used tomitigate a known risk of product con-tamination, usually supported by astrong GMP driver and process data. Itis the foundation of the basic organiza-tion and operation of the facility andidentifies process steps at risk.

Secondary Segregation - usedwhen there is little demonstrated riskto product, but segregation is desirableto minimize the risk of human error andmix-ups. There is no direct impact onproduct quality, but it helps define thefacility and its operation although morefrom a management standpoint than

Page 20: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

intrinsic process protection. Secondarysegregation is open to interpretation asto applications and methodology.

1.3.4.2 Flow and TrafficPatterns in the FacilityImplementation of the segregationstrategies results in “flow.”• Flow patterns should address scale,

volume, and duration of expectedtraffic.

• Flow patterns also should addressupset conditions (such as mainte-nance and change out of large equip-ment) and future construction.

• A mature materials handling phi-losophy must be in place before es-tablishing flow patterns.

Philosophies of primary and secondarysegregation affect flow patterns:• Raw Materials Flow• Product Flow, including intermedi-

ates and hold points• Personnel Flow• Glass and Equipment Flow (through

cleaning protocols)• Waste FlowFlow patterns may force issues withthe cleanliness of the facility:• Materials of construction and archi-

tectural details• Building layout and potential con-

tamination routes (via air, people,equipment)

• Issues with cleaning of equipmentand piping:- CIP/SIP- Wash Facilities

1.3.5 Openversus Closed ProcessingIf a unit operation is demonstratedclosed, it may operate in ControlledNon-Classified (CNC) space.• Closed - segregation by physical

means (hardware) to protect theproduct and process from contami-nation by the surrounding environ-ment (outside the process)

• “Closure” and its measures must bedefined by the Owner, and demon-strated to prevent contamination ofthe product.

• Various operating systems havevarying degrees of closure, some maybe absolute, while others also pro-

vide segregation, but to a lesser de-gree. The use of a “hard” definitionmay limit the understanding of“closed.”

• The surrounding room environmentis not part of the equation for aclosed process, but it should be “con-trolled.”

1.3.5.2 If a unit operation isopen, the product must beprotected by other means.• Open = not “closed”• Product = process + facility• Surrounding environment is a fac-

tor in the process.Either a classified space or a controllednon-classified environment will likelybe needed, but the need for area moni-toring is driven by the open process.

The choice between closed process-ing in Controlled Non-Classified (CNC)space and open processing in classifiedspace is often driven by scale of theprocess, cost of operations, and value ofproduct at risk.

Chapter 4 provides information tohelp in selecting process equipment tomeet open or closed requirements.Chapter 6 discusses the effects of pro-cess closure on the facility.

1.3.6 Scale Affects DecisionsChapters 3, 4, and 5 deal with processdesign and support utility design is-sues connected with process scale, andChapter 6 covers facility layout op-tions.

One size does not fit all. As scale ofthe process increases, there is a shifttoward:• Vertical layouts with gravity flow of

materials• More closed operations• More Primary segregation• Equipment fixed in place (often dedi-

cated)• More automation• Controlled non-classified space in-

stead of classified space (due toclosed processing)

Small process scales tend to include:• Horizontal process flow with pumps• Open operations• Segregation by time (campaigning)• Manual operations (mixing, etc.)• Less automation

• More portable equipment, oftenshared with other products

• More need for classified spaces• Single product vs. multiple prod-

ucts manufactureAs discussed in Chapter 3, when morethan one product is manufactured in afacility, ensuring the products’ safetyand quality becomes more difficult, butno less important. Multi-product manu-facturing facilities may segregate prod-ucts by campaigning (one product at atime) or may process multiple productsconcurrently.• Campaigning depends heavily on

validated cleaning and changeoverprocedures (Chapter 3).

Concurrent manufacturing must avoidcross-contamination through physicalsegregation and operating procedures.(Chapters 3 and 6)

1.4 Using the Guide1.4.1 Organization of the GuideAn overview of the Guide’s structure isshown in Figure 1.

1.4.2 Application of the GuideAs shown in Figure 1, it is necessary tobegin by understanding the GMP re-quirements (Chapter 2) and then ad-dressing the product and operationalrequirements (Chapter 3). From there,once operational concepts have beenestablished, User Requirements de-fined, and perhaps even a FunctionalDesign created, the discipline design-ers may begin detail design.

Users of this Guide are advised torefer to other ISPE Baseline® Guidesfor more detailed or complementaryinformation. For example, water andsteam systems are thoroughly dis-cussed in the ISPE Baseline® Guide forWater and Steam Systems, and thedesign of classified pharmaceuticalmanufacturing space is discussed atlength in the ISPE Baseline® Guide forSterile Manufacturing Facilities.

Users of this Guide are also encour-aged to understand GMP and specificproduct requirements thoroughly be-fore attempting facility design. Wherethere is conflict or a lack of under-standing, manufacturers and engineersare encouraged to discuss concepts withthe appropriate regulatory agency.

Page 21: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

Such early discussion opens dialogueand helps to settle potentially thornyissues.

2. The Regulatory Basis forFacility Requirements

During the design of new facilities,every manufacturer faces numerousissues that may significantly affect thefacility cost. These include process defi-nition, process equipment require-ments, the definition of a suitablemanufacturing environmental qualityto support manufacturing, water re-quirements, and facility layout. Whilesome of the issues faced may affect thequality of the Active PharmaceuticalIngredient (API or bulk drug sub-stance), others may have no impact.

The primary element to be consid-ered in a biopharmaceutical facility isthe ability of the facility and the pro-cess to protect, i.e., prevent contamina-tion of, the API. Product protectionissues may be addressed in a voluntaryProduct Protection Control Strategy.

The evolution of facilities for manu-facturing biopharmaceutical productshas led to many extremes in size, com-plexity, and capital/resources. Process-ing approaches and designs suitablefor a small-scale process are often in-adequate or inappropriate for a large-scale facility. The multi-product facil-ity will differ in certain key areas fromeither of these dedicated facilities.

Specifically, each company shoulddetermine the appropriate require-ments to provide adequate protectionfor its product(s), and thereby, the re-quirements for the completed facility.No single solution or design fits alldrug substances or products since thedecisions made and incorporated inthe facility will depend upon:• Nature of the process and product

(i.e., contamination-sensitive pro-cesses to less sensitive processes,open versus closed processing, etc.)

• Scale and complexity of the process• Number and types of the products

in the facilityThis Chapter addresses some of thesignificant process-related conceptsand facility attributes with regulatoryimplications to be considered whendesigning a facility. Key points devel-

oped include:• There is not one universal “GMP”

standard or approach to biophar-maceutical facility and process de-sign. The nature of the product andits processes greatly influences thesedecisions. Systems and their com-ponents will have varying effect oneach product.

• Biotech manufacturing operationsare not usually intended to producea sterile drug substance, but ratherone of low bioburden. Although thevoluntary adoption of aseptic manu-facturing techniques and facilitystandards have occurred in the in-dustry, such standards are not re-quired. The production process andfacility should include the appropri-ate controls to prevent, limit, anddetect API contamination.

• Processes may be closed or open.Closed processing presents less riskto product and presents fewer de-mands on the facility design. Localcontrols may be used with open pro-cesses to provide protection of theproduct.

• Multiple products segregated byappropriate procedural or physicalmeans may be produced within asingle facility.

• Water used in manufacture shouldbe appropriate to the process; WFImay not be scientifically necessarythroughout the entire process formost products.

3. ManufacturingOperations and Activities

This Chapter involves the operationalaspects of a biopharmaceutical facil-ity, as opposed to the physical design ofthe facility itself, and addresses keyregulatory issues and concepts definedin Chapter 2. The Chapter addressesthe impact of facility and equipmentdesign decisions on manufacturing op-erations. Conversely, the Chapter alsodescribes how operability and main-tainability considerations should in-fluence the design of a biophar-maceutical facility. Concerns and is-sues of production management, pro-cess operators, and other plant supportpersonnel are included. Important con-cepts addressed in this Chapter are as

follows:• Operational and Procedural

Controls can play an importantrole in protecting the product, andmust be factored into the “open ver-sus closed” design decision. Appli-cation of these types of controls witha well trained manufacturing staffcan often be a better solution thanover-engineering a system.

• “Bioburden-Controlled Process-ing” and “Pyrogen (Endotoxin)-Controlled Processing” are keyoperational concepts that have asignificant impact on process andfacility design, and both are dis-tinctly different from sterile pro-cessing. Some features of traditionalsterile design and operation may beemployed, but are typically not re-quired to establish the appropriatelevel of control.

• Viral Clearance (Reduction andInactivation) – Biopharmaceuticalprocesses commonly use raw mate-rials from biological sources, start-ing with the cell line and often ex-tending to supplements added dur-ing the cell culture and purificationstages. Cell lines used in the bio-technology industry are extensivelycharacterized for identity and pu-rity, and are tested for the presenceof infectious agents. Nevertheless,it is still a regulatory requirementfor manufacturers using mamma-lian cell culture-based processes todemonstrate adequate viral clear-ance during the manufacturing pro-cess. In addition, increasing con-cern over the transmission of prionsfrom animal-sourced raw materialshas required manufacturers to takeadditional measures to minimize therisk of such contamination. The de-cision on how/where to accomplishviral clearance can have an impacton the equipment design, and mayaffect the design and layout of thefacility.

• Segregation is critical in anybiopharmaceutical operation to en-sure product protection. Traditionalapplications include:- Between organisms, products, or

technologies- Between processing steps (e.g.,

Page 22: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

upstream and downstream op-erations)

- Between raw materials or prod-ucts at various stages of qualitycontrol or process step

- Between components or equip-ment at different stages of clean-liness

Segregation can be accomplished byprocedure, by spatial separation(physical), by time (temporal), byenvironmental control, or by pro-cess design (system closure).

• In a Multi-Product Operation,products can be either campaignedor processed concurrently. For cam-paigned products, the focus is oncleaning validation, changeover pro-cedures between products, and lineclearance procedures. For concur-rent product manufacture, the fo-cus is on segregation, proceduralcontrols, and avoidance of cross-con-tamination. In all cases, the overallguiding principle is to ensure thequality and safety of the product.

• Manufacturing at DifferentStages of Product Developmentis important for many biophar-maceutical companies, particularlythose facing their first major capitalinvestment in manufacturing facili-ties. While the regulations are clearin stating that GMP compliance isrequired for all stages of clinicaldevelopment, it is also recognizedthat in most cases the manufactur-ing process is not completely de-fined during early-stage clinicalwork. It is important that processissues having significant impact onthe facility design be locked down asearly as possible. The emphasis ofprocess/facility design and valida-tion during early-stage clinicalmanufacturing should be placed onareas that have the greatest impacton product quality and consistency.

4. Process and EquipmentThe Chapter on Process and Equip-ment is primarily concerned with de-sign aspects of biopharmaceutical pro-cesses and equipment, as opposed to therelated operational aspects addressedin Chapter 3. More specifically, thisChapter deals with the design of

biopharmaceutical process equipment,and associated piping and instrumen-tation, which contact a product or itscomponents at a stage in the processwhere such contact could influence thequality, safety, purity, strength, or iden-tity of the ultimate product. The pri-mary audience for this Chapter is pro-cess and equipment engineers.

In general, biopharmaceutical pro-cesses are similar in that nearly allhave fermentation/cell culture produc-tion steps, harvest steps, purificationsteps, formulation steps, and final bulkfilling steps. Although manufacturingprocesses may differ, certain CriticalProcess Parameters are consistent fromproduct to product, and certain keyconsiderations for each processing stepapply to all processes.

Within each process step, there areprocess considerations driven by theoverall philosophy of the organizationoperating the process. The design ap-proach that is chosen based on theseconsiderations (GMP and business driv-ers) will result in a set of criteria to beused for both equipment selection andoverall facility design. There is no singleanswer to the majority of the processconsiderations mentioned. However,the combinations of the choices andsolutions will define reasonable, com-pliant process designs.

Various types of equipment sharesimilar design considerations and re-quirements. Specifically, cleanability/drainability, surface finish, materialsof construction, shear generation, clo-sure level, containment level, and pres-sure/temperature requirements mustbe considered for virtually any piece ofequipment or device used in biologicalmanufacturing. Improper consider-ation can lead to processing systemsthat are either not operable (placingproduct at risk) or are operationallyinefficient (lower process yields).

Key topics addressed in this Chap-ter are:• Simplified process flow diagrams of

several typical biopharma-ceutical processes.

• Critical process parameters thatare consistent from product to prod-uct. Key processing (critical) param-eters for various processing steps

are identified for typical unit opera-tions. Critical process parameters,such as temperature, pH, conduc-tivity, bioburden, endotoxin, prod-uct concentration, by-product lev-els, purity, and stability are gener-ally similar from process to process.However, the acceptance criteria,implications, and applicable designoptions from process to process mayvary significantly.

• General considerations forequipment design – design con-siderations common to mostbiopharmaceutical unit operations.

• General equipment consider-ations, such as materials of con-struction, cleanability, avoidingcross contamination, open vs. closed,process monitoring, safety, contain-ment, and maintenance, can be ap-plied to most process equipment,and design considerations are out-lined. Similarly, there are designconsiderations applying specificallyto general particular areas such ascell culture and purification. Theseare outlined as well in the form ofchecklists for the process and equip-ment engineer.

• Specific equipment design con-siderations – design considerationsthat are unique to certain specificbiopharmaceutical process equip-ment types.

Although a detailed analysis ofevery unit operation used in biophar-maceutical processes cannot be cov-ered in this Guide, unit operationsgenerally fall within these broadprocess operation areas:- Raw Material Storage/Handling- Weigh/Dispense- Media/Buffer/Component Prepa-

ration/Hold- Inoculum Preparation- Fermentation/Cell Culture- Recovery/Harvest- Purification- Bulk Filling- CIP- SIP- Biowaste Deactivation

Specific design issues affecting unitoperations in these areas are outlined.

Page 23: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

5. ProcessSupport and Utilities

This Chapter provides guidance in de-sign and operation of utility servicessupporting the manufacturing ofbiopharmaceutical products. Utilitysystems addressed in the Chapter in-clude:• Pharmaceutical Water Systems• Cleaning, Sterilization, and Depyro-

genation Systems• Process and Utility Gases• Process Temperature Control Sys-

tems• Biowaste and Process Waste Han-

dling• Seal Support Systems• Plumbing and Piping Systems• Emergency PowerThe Chapter focuses on process sup-port systems that affect ability to meetGMP production requirements. TheChapter identifies the major GMP is-sues for each of the systems addressed.Guidance is provided in design of sys-tems to minimize risks of product con-tamination or unreliable production.

For purposes of qualification andcommissioning, the Chapter categorizesprocess support utilities as having “Di-rect Impact,” “Indirect Impact,” and “NoImpact” on product. The Chapter rec-ommends full qualification and com-missioning of Direct Impact systems.Systems with Indirect Impact or NoImpact should be commissioned consis-tent with Good Engineering Practice.

Key Conceptsdiscussed in this Chapter• Process support system features that

affect GMP are identified, and vul-nerable characteristics are ex-plained

• Methods to minimize product con-tamination risks from process sup-port utility systems are presented

• Except when required for safety oroperational reasons, system designshould minimize the need to serviceand otherwise access process sup-port systems from within produc-tion areas

• Systems that might enable trans-mission of contaminants are identi-fied with methods for preventionprovided

• Methods to define commissioningand qualification requirements forprocess support utilities

• A summary of key concepts forbiopharmaceutical water systemsis provided

6. FacilityBiopharmaceutical manufacturing fa-cilities are very complex and resultfrom projects that focus on the at-tributes of the product(s) being pro-duced, the attributes of the process,and the facility attributes needed tomeet cGMP guidelines. The facilitydesign team should become familiarwith the topics discussed in the Chap-ter to understand how each will affectthe final facility design and operation.

This Chapter will review:• The impacts of process and unit

operations on facility design• How product attributes play a key

role in facility definition• The importance of adjacencies in

defining operational flow to mini-mize potential contamination op-portunities

• The impacts of containment andclosed processing on facility design

• The definition of area environmentsand their impact on facility layoutand design

• The issues related to single productvs. multi product production phi-losophy

• Air lock and gowning room alterna-tives

• Considerations for effective processand production support areas

• Regulatory considerations in facil-ity design

• Layout alternatives, when is verti-cal flow practical

• Finishes are covered in otherBaseline® Guides, and are refer-enced in this Chapter

• Discretionary (non-GMP) consider-ations

7. Process ControlsThis Chapter on Process Controls andAutomation provides points to considerwhen developing instrumentation andautomation strategies forBiopharmaceutical operations. Thisprocess starts by determining the de-

tails of the biological process to becontrolled. What are the critical oper-ating conditions? What can adverselyaffect the process or product? Once theprocess and critical operating param-eters are identified, the optimal levelof automation versus control viamanual procedures may be determined.

Automation is not a GMP require-ment. However, when automation isused, it carries with it GMP require-ments. If properly applied and vali-dated, automation can help achieveongoing GMP compliance. When notproperly managed and designed, auto-mation can result in problems withproject schedule and cost.

Topics covered in this Chapter areorganized as follows:• Biopharmaceutical Automation Is-

sues• Level of Automation• Biopharmaceutical Unit Operations

- Fermentation/Cell Culture- Cross Flow Filtration- Chromatography- SIP- CIP

• Control System Maintenance• Validation of Automation Systems

8. Commissioningand Qualification

A biopharmaceutical manufacturing fa-cility is commissioned and qualified inthe same manner as any other pharma-ceutical manufacturing facility. Manyaspects of the qualification of asepticmanufacturing facilities apply to classi-fied spaces in biopharmaceutical facili-ties, yet there are many areas that re-quire only commissioning in accordancewith Good Engineering Practice.

It is imperative that, before detaildesign begins, the owner and engi-neers develop User Requirements(What the facility is to do) and a Func-tional Design (How the facility willwork and protect the product). Theseactivities will identify product/processcritical parameters and their accep-tance criteria (forward processing cri-teria), against which post-constructionqualification will verify performanceof the direct impact systems that areidentified in Functional Design.

Page 24: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Biopharmaceuticals

8 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

The ISPE Baseline® Guide for Com-missioning and Qualification providesvaluable guidance in identifying thesystems needing qualification. Ratherthan restating the entire Guide, a fewhighlights are provided in this Chap-ter. The facility engineer is directed tothe ISPE Baseline® Guide for Commis-sioning and Qualification for furtherinformation.

9. GlossaryA glossary of pharmaceutical industryterminology relevant to the ISPEBaseline® Guide for Biopharma-ceuticals.

10. Appendix -European Aspects

The purpose of this appendix is to high-light the general requirements in Eu-rope and to point out the differencesbetween Europe and the US. WithinEurope, the majority of countries arecovered by the European Union (EU).There are countries in Europe outsideEU, such as Switzerland, that are cov-ered by their own national regulations.The general trend is to harmonize theregulatory requirements worldwide,but differences still exist. Organiza-tions like ICH are the main drivers forthat development.

Within Europe, the EU directivesare harmonizing general requirements,giving the minimum standards. Thenational laws need to comply withthese, but are allowed to be more strin-gent.

Page 25: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

Design Criteria and Evaluation ofPharmaceutical Containment SystemsEvaluation for Open Isolation Systemsby Osamu Suzuki, PhD, Morihiko Takeda, Koji Tanaka,and Mikio Inoue, PhD

This articledescribes designcriteria as astrategy forcontainmentsystems(isolationsystems)applicable topharmaceuticalfacilitieshandling ActivePharmaceuticalIngredients(APIs).

Introduction

Recent drug development is becomingprogressively directed toward drug po-tency.1-4 For the present, this tendencyis likely to continue, increasingly fa-

cilitated by the advancement of the develop-ment of drugs targeting biochemical pathways,cellular controls or gene regulators, associatedwith specific genes, and based on genetic re-search and cell biology.3 This trend contributesto approvals for reductions in patient drugdosage with a concurrent reduction in the al-lowable exposure level to the drug componentfor the workers during the manufacturing pro-cess.4 Pharmaceutical containment systems(isolation systems) allow pharmaceutical manu-

facturers to meet hazardous chemical materi-als exposure limits and are an essential part ofpharmaceutical current Good ManufacturingPractice (cGMP).4 This technology provides forthe design of unique pharmaceutical contain-ment facilities, including closed and open isola-tion systems within aseptic2,5-10 or non-aseptic1-

3,6,7,9-10 environments. Pharmaceutical companies,engineering contractors, and equipment sup-pliers have recently, but independently, madean effort to establish exposure control limitsthrough conceptual classification or case stud-ies with various surrogate materials.4,11-15 Thisarticle proposes design criteria to assist in thecontainment of potent compounds16-20 and re-flects these criteria in the design of pharmaceu-

tical bulk and finished product manu-facturing facilities. The laboratory un-der study has recently joined a researchproject to evaluate the containment per-formances of APIs in pharmaceuticalfacilities. The purpose of the researchproject is:

• to investigate quantitatively the con-tainment level of the APIs to confirmthat containment performances metdesign criteria

• to establish the quantitative designcriteria based on the analyses of thebehavior of the airborne dust in po-tent compounds within manufactur-ing processes

In the study, the containment perfor-mances of an open-type containmentisolation system, which could be catego-rized to a Performance-Based ExposureControl Limit (PB-ECL)11 of 3 were in-

Figure 1. Schematicview of an opencontainment cabinet.

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 26: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

vestigated. The review report7 focuses on highlights in the NewPDA Technical Report No.34, which defines the open isolatorsthat are designed to allow for the semi-continuous egress ofmaterials during operation while maintaining a level of protec-tion over the internal environment. The isolation systemsevaluated in the study included both a closed isolator (glovebox) and a containment cabinet of safety cabinet-type (opencontainment cabinet). The open containment cabinet is de-signed to provide a flow of filtered air over the product, whileensuring that the flow of air is into the cabinet. This type ofcabinet also ensures efficient working, while maintaining alevel of protection over the internal environment.21 Attentionwas focused on illustrating the relationship between theworker’s operation and the containment performances underAPI processing in bulk API manufacturing facilities.

Design Criteria forPharmaceutical Facilities

The general strategy of the facility design for the pharmaceu-tical containment systems is as follows:16,19

• Classification of bulk or finished pharmaceutical productsinto five hazard categories (PB-ECL)11 according to theirinherent toxicological and pharmacological properties,shown in Table A.

This PB-ECL classification is based on an earlier report11 andpast practices of pharmaceutical facilities in containing thevarious potent compounds. The categorization by PB-ECL foreach pharmacological property, as the finished product, isshown in Table A.

• Classification of the barrier level into classes from zero to2.0, at 0.5 intervals, by integrating various containmentsystems, including personal protection, is shown in Table B.

The protection systems for the external environment, such aslayout zoning, HVAC system, and building construction, alsoare considered.

• Selection of the barrier level to maintain the requiredenvironment for each manufacturing process.

The barrier level should be selected according to the amountof dust generated by conditions, such as water content andhandling volume, shown in Table C.

• Integrated study to reflect the selected barrier level in thefacility design by utilizing a combination of barrier tech-nologies, gowning regulations, and the layout of buildingfacilities.

Containment Strategy and Facilities toProcess Bulk APIs

Pharmacological Properties of the DrugThe categorization by PB-ECL for each pharmacologicalproperty, as the finished product, is shown in Table A.Although the intravenous toxicity was assigned to an ECLcategory of 3, other pharmacological properties includingcarcinogenicity, sensitivity, and pharmaceutical potency maybe assigned to an ECL category of 2. However, as the intrave-nous toxicity was considered as the critical point, ECL cat-egory 3 was selected as the design basis for bulk manufactur-ing facilities.

Table A. Performance-based exposure control limit (PB-ECL)11 for pharmaceutical drug manufacturing.

: ECL category in each pharmacological property as finished product

PB-ECL Category 1 2 3 4 5

Exposure Level (µ g/m3) 1000~5000 100~1000 1~100 <1 NIL

1. Active Potency (mg/day) >100 10-100 0.1-10 <0.1 <0.1

2. Hazard Toxicity LD50 (mg/kgRat) >2000: 500-2000: almost 50-500: slightly toxic 5-50: toxic <5: highly toxicToxicity of Oral non toxic non toxicOSHA/HCS >500: non toxic 50-500: toxic <50: highly toxicWHMIS (Canada) >500: non toxic 50-500: toxic <50: highly toxicToxic Control Law >300: non toxic 30-300: slightly toxic <30: toxicOcean Pollution >2000: non-hazardous 500-2000: practically 50-500: slightly 5-50: moderately <5: highly hazard Control (GESAMP) non hazardous hazardous hazardous

Toxicity of >100: non toxic 7-100: toxic <7: highly toxic intravenous

3. Others Carcinogenicity (IARC) ----- ----- ----- 2A, 2B: potentially yes 1: yes----- ----- -----

Sensitivity low low-middle middle middle-high high

Page 27: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

Barrier Level Settingfor each Manufacturing ProcessTable D shows the required barrier level setting for eachmanufacturing process. The barrier level is defined, accord-ing to the condition of the bulk APIs, with the values of thelevel varying between 0 and 2.0,16-20 as shown in Table C.

Table B. Barrier level setting for worker protection.

Barrier level Definition for protection of worker andenvironment against potent compounds

0 Not protected

0.5 Partially protected

1.0 Fully protected

1.5 More protected

2.0 Doubly protected

The process for manufacturing the bulk APIs includesweighing of raw material within the glove box, charging of theraw material into a reaction vessel (suspending of the rawmaterial), crystallization, filtration, cake receiving and dry-ing, along with weighing, milling, and dispensing within theopen containment cabinet, with further mixing, and finally,dispensing for packaging as a bulk product.

Crystallization, filtration, cake receiving, and drying pro-cesses were given a barrier level of 1.0, and with a value of 1.5selected as the required barrier level for the weighing withinthe glove box charging, the powder handling processes withinthe open containment cabinet, and subsequent mixing anddispensing processes are considered the highest levels in thewhole handling processes. This barrier level value corre-sponds to the handling of a large amount of powder. Thecontainment performances were evaluated for the weighingprocess in the glove box, the subsequent charging process intothe reaction vessel, and the powder handling processes in theopen containment cabinet.

Powder HandlingProcesses in Containment SystemsThe raw material was taken from the bag within the glove boxand subdivided by weight. The accurately weighed raw mate-rial was packaged into small bags. Each bag was taken from

Table C. Barrier level according to the condition of hazardouschemicals under ECL.

Barrier Level

Condition Exposure Control Limit

1 2 3 4 5

Large amount of powder 2.0

Small amount of powder1.0 1.5

1.5 ≥Liquid/wet powder 0.5 1.0 1.0 2.0

Very small amount of 0.50.5 0.5powder/liquid

Powder/liquid enclosed 0 0 0 0 0.5

Non-hazardous substances 0 0 0 0 0

Figure 2. Schematic view of a mock-up booth representing aturbulent flow booth.

Table D. Process flow and barrier level setting.

Process Flow Required Barrier level

Weighing of raw material 1.5

Charging of raw material 1.5

CrystallizationFiltration 1.0Cake ReceivingDrying

Weighing/Milling/Dispensing 1.5

Mixing 1.5

Dispensing 1.5

the glove box through the pass box and placed in a closedpowder loading system. The system was then connected to theinlet of the reaction vessel for the material charging. After theinstallation of this system, the raw material was thoroughlycharged into the reaction vessel for the subsequent crystalliza-tion. The inside of the bag was well cleaned by purified water,which was introduced from a nozzle installed inside the vessel,leaving the bag ‘as is,’ and the loading system was uninstalled.After filtration and drying, the powder was handled on adifferent day for weighing, milling, and dispensing within theopen containment cabinet. The equipment surfaces and floorswere cleaned for both processes soon after the operations werecompleted. The containment evaluation was performed subse-quent to the operations, but before the cleaning. Figure 1illustrates the lateral view of the open containment cabinet.Because this cabinet is open booth, the inside is at atmosphericpressure. However, this cabinet is designed to give a flow offiltered air over the product while ensuring that the flow of airis into the cabinet. This type of cabinet also ensures efficientworking, while maintaining a level of protection over theinternal environment.21

Page 28: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 4. Interior surface contaminant concentrations of a glovebox after the raw material weighing process.

swabbing. A previous study using this swab method confirmeda recovery rate of 95% confidence of the surrogate materialfrom the surface although an organic solvent was used fordipping swab clothes in this earlier study.22

Mock-up TestParameters for the Test and Mock-up BoothThe mock-up test covers several parameters that should beconsidered in the design of isolation systems. They includestudies of the isolation systems, such as whether it is closedor open; flow patterns, whether it is laminar or turbulent;efficacy of local exhaust ventilation; different types of thesurrogate materials and the particle size distributions, andtype of sampler head, such as the cassette type closed head orthe IOM head for air sampling.

Figure 2 shows a schematic view of the mock-up booth,which represents a turbulent flow booth with a local exhaust.A dust feeder was used to secure constant feeding of thesurrogate material. Since the study focuses on the contain-ment evaluation of the open isolation system, the result of thecontainment performance for the open booth with turbulentflow in line with the parameters listed is presented with onesurrogate material. The cassette type closed head was usedfor air sampling. Monitoring times for air sampling werebetween 20 and 30 minutes. Temperature and relative hu-midity during the measurements were 20ºC and 38%, respec-tively.

Surrogate MaterialFine lactose was used as the surrogate (test) material with aparticle size range by weight ratio (provided by the manufac-turers) of:

• below 50 µm (>30%)• 50µm to 75µm (<35%)• 75 µm to 150 µm (<25%)• over 150 µm (<10%)

Evaluation MethodsSampling Methods for Containment Evaluation

Air SamplingConstant flow air samplers, using a 37 mm diameter, cassettetype closed head or the International Occupational Medicine(IOM) head mounting and adequate filter, were used tomonitor the concentrations of the airborne dust from powderhandling during operations.

Static or personal monitoring was carried out. The moni-toring commenced just before operations commenced andconcluded just after operations were completed. The Short-Term Particulate Airborne Concentration (STPAC) was de-termined for less than one hour of operation, which was theusual amount of time required to complete the operations,and measured as units of amount collected per unit volume.The cassette type closed head was normally used, except inthe measurement of the raw material charging into thereaction vessel during a comparison with the IOM head forperformance.

Swab TestInterior and/or exterior surfaces of equipment, surfaces offloors in the work place, and operator gowns were investigatedto determine the degree of surface contamination (total amountrecovered or amount per unit surface area) by a validated swabmethod.22 Cloth was dipped in purified water and then squeezed.It was used for swabbing. Particular attention was focused onthe entry and exit of workers from the workroom with respectto any probable carry-over of the powders being processed. Forthis purpose, the swab test was performed for those floors thatwere traversed by the operators.

Analytical MethodThe concentrations of the airborne and the surface-contami-nated particulate were analyzed by a validated chemicalanalysis. The recovery rates from both the filter and the clothwere validated and reflected in the analytical results. Whilethe recovery rate from the surface was not determined becauseof the restriction of the use of water as solvent, the surface wasvisually checked to confirm that no powder remained after

Figure 3. Containment evaluation by air sampling for inside andoutside a glove box after the raw material weighing process.

Page 29: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

Results and DiscussionEvaluation of Containment Performancesfor Weighing and Charging of Raw Materials

Glove Box and its Surrounding Environmentduring WeighingFigure 3 shows the sampling point and the analytical resultsfor air sampling. Air sampling was carried out using thecassette type closed head. This figure is illustrated in planview. The pressure within the pass box was designed to be

Figure 5. The sampling points and the analytical results for bothair sampling and swab test of the operator gown during thecharging of the raw material.

negative with respect to the atmospheric pressure of theworkplace. The pressure within the glove box was negative tothat of the pass box. The containment performance of theglove box was estimated by air sampling:

• inside the glove box• inside the pass box• in front of (outside) the pass box• in front of the exhaust of the room

In order to confirm the performance of the HEPA filter forpossible inclusion of sub-micron particles, air sampling wascarried out at the outlet side of the HEPA filter of the glovebox. The sampling in the case inside the glove box was carriedout in front of the exhaust (HEPA filter) to allow the resultsto be compared before and after the HEPA filter. The sam-pling for the outlet of the HEPA filter was carried out usinga port for measuring the differential pressure. The analyticalresults demonstrated that the STPAC in the glove box andthe pass box were 298 and 35 µg/m3, respectively, while theenvironment outside the glove box, including the outlet of theHEPA filter, were shown to be under the detectable limit (<1 µg/m3). These results demonstrated that the raw materialhandled was satisfactorily contained within the containmentequipment (the glove box) and that the containment perfor-mance met the design criteria. The weighing process wascompleted within 10 minutes. The airborne dust of the raw

Figure 6. Containment evaluation relevant to worker operation by swab test after weighing and charging processes.

Page 30: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

evaluated along the exit route from the workplaces after theweighing and the charging processes shown in Figures 3-5.Swab sites with an area of 0.25cm2 (50cm × 50cm) werepredetermined and the sites were swabbed before startingthe operations. After the operations and subsequent cleaningof the floors around the workplace, the corresponding siteswere swabbed again to determine the surface concentrations.The surface concentrations shown in Figure 6 were expressedas the difference between these two swab tests.

The results revealed that the workplace surfaces werecontaminated, but concentrations were markedly decreasedbefore entering the gowning room, which is a space fordegowning (from 86 µg/0.25m2 to 46 µg/0.25m2). However,contamination was still detected in the gowning room (10 µg/0.25m2), in the degowning room (1.5 µg/0.25m2), and in aspace for putting on shoes (1.1 µg/0.25m2); although theconcentrations were below the detectable limit after thedegowning room (<1 µg/0.25m2). This provided clarificationthat the powder handled was fully contained within thehazardous areas. However, these results suggest that thereis a possibility of carry-over of the handled powder outside theworkplace, most probably on workers’ gowns.

These results also indicate that the carry-over by workers,following exposure to the airborne dust of the powder handled,needs to be considered in the design of the pharmaceuticalcontainment facilities, in addition to the containment ofairborne dust.

Evaluation of Containment Performancesof Open Isolation Systems

Surrounding Environment of OpenContainment Cabinet during Powder HandlingFigure 7 shows the sampling points and the analytical resultsfor both the air sampling and swab test in the powderhandling processes within the open containment cabinet. Theprocesses include the weighing, milling, and dispensing ofthe re-crystallized active compound. All processes were car-ried out within approximately 1.5 hours. Air sampling wascarried out during these processes. The STPAC inside thecabinet was estimated at 88 µg/m3 where the weighing anddispensing processes were performed, whereas it was only 5.1µg/m3 outside the cabinet and 2.6 µg/m3 in front of the exhaustof the room. The STPACs were 4.2 µg/m3 next to the millinside the cabinet and 5.9 µg/m3 outside the cabinet. Theseresults indicate that the airborne dust was primarily pro-duced by the weighing and dispensing processes, rather thanthe milling process. The results also suggest that the opencontainment cabinet has the ability to contain the powderhandled. The floor surface concentrations in front of thecontainment cabinet were estimated as 86 mg for the weigh-ing process and 19 mg for the milling process. The reason thatthe concentrations were expressed as an absolute amountrecovered was that the powders collected by the swab testwere easily visible as masses on the floor surfaces close to theopen containment cabinet, in both cases.

The overall results seem to confirm a satisfactory perfor-mance of the open containment cabinet to contain relatively

material also was found within the pass box. This might becaused by moving the bag from the glove box to the pass boxrelatively soon after the weighing process, despite the differ-ential pressure between the glove box and the pass box. Thisemphasizes the importance of using the exhaust in the glovebox effectively, by taking sufficient time to remove the air-borne dust.

Figure 4 shows the analytical results for the interiorsurface concentrations at the front and the lateral sideswithin the glove box after weighing. The concentration of thelateral side close to the exhaust was obtained by swabbing theneighboring site of the exhaust. The interior surface concen-trations were shown to increase toward the lower side fromthe upper side, suggesting this as a pattern of behavior of theairborne dust within the glove box with regard to the particleconcentration of the raw material during weighing.

Surrounding Environmentof Reaction Vessel during ChargingFigure 5 shows the sampling points and the analytical resultsfor both air sampling and swab test of the operator gownduring the charging of the raw material. The performance ofthe cassette head was compared to the IOM head in airsampling. These sampling heads were positioned at a dis-tance 200 mm from the inlet edge of the reaction vessel, byfacing the sampling heads toward the lateral side of the inlet,perpendicularly. The STPACs were estimated as 92µg/m3 forthe cassette head and 77µg/m3 for the IOM head, indicatingrelatively compatible sampling performances for collectingthe airborne dust of the raw materials. These STPACs weremuch lower than that within the glove box and within therange of the assumed design criteria. The STPACs corre-sponded to 4 to 5µg/m3 when they were expressed by 8 hourstime weighted average. Because the material charging pro-cess was completed within 26 minutes, the existence of theairborne dust seems to be instantaneous. It is likely that therelease of the airborne dust from the inlet of the reactionvessel is the result of disconnecting the closed powder loadingsystem relatively soon after charging the raw material and/or insufficient cleaning by purified water for the small amountof the residue within the bag. This implies that it is highlypossible to decrease the STPACs by improving operationprocedures. The release of the airborne dust caused simulta-neous exposure of the operator gown, as shown in Figure 5.The surface concentration was found to be 36 µg/625cm2. Thearea of 625cm2 (25 cm × 25 cm) corresponded to the directswab area. Although the amount of the raw material detectedin the operator gown was small, the containment evaluationduring the charging process strongly suggests the impor-tance of gowning as a secondary barrier, and of consideringadequate regulation for gowning during the facility design.

Containment PerformancesRelevant to Worker OperationsParticular attention was given to the containment perfor-mances associated with the operation of workers. Figure 6shows the floor surface concentrations of the raw material

Page 31: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

Figure 8. Containment evaluation by air sampling (personal monitoring)and swab test for an operator gown after weighing, milling, anddispensing processes within anopen containment cabinet.

large amounts of the powder being handled with a betteroperational performance when compared with the closedsystems, such as the glove box. The best type of containmentsystem can be adequately selected by considering both theoperational performance and the PB-ECL for the potentcompounds.

The release of the small amount of airborne dust from theopen containment cabinet, however, caused the exposure tothe operators’ gown. Figure 8 shows the analytical results forboth personal air sampling and the swab test of the gownafter the powder handling within the open containmentcabinet. The STPAC obtained by the personal monitoringwas only 20 µg/m3, which corresponded to 3 µg/m3 by 8 hourstime weighted average for 75 minutes of operation. The gownsurface concentration was estimated as 250 µg/625cm2, which

Figure 7. Containment evaluation by air sampling for an opencontainment cabinet and swab test for floors during weighing, millingand dispensing processes within an open containment cabinet.

was higher than that of the charging process of the rawmaterial. This may result from the sustained exposure in thissituation (75 minutes) compared to the charging process (26minutes).

A similar tendency was obtained for the evaluation of thefloor surface contaminations to the workplace exit by opera-tors, after the powder handling of the open containmentcabinet (data not included). The results indicate that al-though some carry-over of the powder on the operator gownwas found, complete containment within the hazardous ar-eas was accomplished.

Complementary Effect of the Mock-Up Teston Containment Performance Evaluation

Figure 9 shows the STPACs of the fine lactose for the basecase (no airflow case) and its comparison (turbulent flow andexhaust working case) to the mock-up open booth. The nu-merical values were shown for both inside and outside thebooth. The results demonstrated the quantitative efficacy ofthe airflows to decrease the STPAC (from 30,000 µg/m3 to4,500 µg/m3 for inside and 7,500 µg/m3 to 18 µg/m3 for outsidethe booth). These results also provide a quantitative back-ground for the containment performances of the bulk phar-maceutical facilities evaluated in the present study. Recentcomputational fluid dynamics studies suggest that the ex-haust has an important role to contain the compound by thedesigned airflow in the transfer vessel when having a localexhaust system.8 In such an open isolation system, it appearsthat an understanding of the airflow is critical to ensure thecontainment performance. Conversely, it has been shownthat particle size distributions of airborne dusts differeddepending on the state of the powders, despite being of thesame surrogate materials,23 suggesting the different behav-ior for airborne distribution. Further study at the variousconditions with different surrogate materials is required fora more comprehensive understanding of the containmentperformances of the open isolation systems.

ConclusionFrom the measurement of the containment level of the glovebox and the open containment cabinet (the open isolationsystem), the present study can be summarized as follows:

• The containment performances of the pharmaceuticalfacilities including the open isolation system, designed toa PB-ECL category of 3, were confirmed to meet therequired design criteria.

• The performance of the open isolation system was comple-mented when using the mock-up booth, resulting in anenhanced pharmaceutical facility performance and theability to demonstrate containment performance to re-duce airborne dust.

• The importance of gowning as a secondary barrier wasascertained and it was suggested that adequate regula-tion for gowning during facility design should be consid-

Page 32: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

8 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

ered. The carry-over of the powder on the operators gown,after working, is one of the factors that needs to beconsidered for the containment design, as well as thecontainment of the airborne dust.

• Further evaluations of the pharmaceutical containmentsystems, such as aseptic isolation systems, are under wayto examine the proposed containment facility design crite-ria based on the quantitative analyses in conjunction withthe mock-up test.

References1. Hines, M.J., “Conceptual Design of Isolators for Handling

and Processing Dry Bulk Pharmaceutical Chemicals,”Pharmaceutical Engineering, Vol. 18, No. 2, 1998, pp.60-78.

2. Belly, S., and Wilkins, J., “A Technical Review of Isola-tors,” Pharmaceutical Engineering Vol. 18, No. 2,1998, pp. 80-87.

3. Guest, I., “Industrial Hygiene Challenges Presented bythe Trends in New Chemical Entities,” ISPE Confer-ence, Amsterdam, Netherlands, December 8-9, 1999.

4. Liberman, D., Lockwood, C., McConnell-Meachen, M.,McNally, E., Rahe, H., Shepard, K., and Snow, G., “Bar-rier Isolation Technology: A Safe and Effective Solutionfor Providing Pharmaceutical Development Facilities,”Pharmaceutical Engineering, Vol. 21, No. 4, 2001, pp.22-30.

5. Friedman, R., “Design of Barrier Isolators for AsepticProcessing: A GMP Perspective,” Pharmaceutical En-gineering, Vol. 18, No. 2, 1998, pp. 28-36.

Figure 9. Containment evaluation in a mock-up open booth.

6. Francis, L., “The History and Principles of Barrier Tech-nology for the Pharmaceutical and Biotech Industries,”Pharmaceutical Engineering, Vol. 18, No. 2, 1998, pp.38-47.

7. Madsen ,E.R., “Design and Validation of Isolator Systemsfor the Manufacturing and Testing of Health care Prod-ucts. Highlights from the New PDA Technical ReportNo.34,” Pharmaceutical Technology, Vol. 25, No.2,2001, pp. 40-44.

8. Ramsden, A., “The Use of Airflow Modeling in the Designof Pharmaceutical Containment Systems,” Pharmaceu-tical Engineering, Vol. 21, 2001, No. 4, pp. 40-46.

9. Lysfjord, J., Porter, M., “Barrier Isolation History andTrends, A Millennium Update,” Pharmaceutical Engi-neering, Vol. 21, No. 2, 2001, pp. 142-148.

10. Lysfjord, J., Porter, M., “Barrier Isolation History andTrends,” Pharmaceutical Engineering, Vol. 18, No. 5,1998, 2001, pp. 80-86.

11. Naumann, B.D., Sargent, E.V., Starkman, B.S., Fraser,W.J., Becher, G.T., and Kirk, G.D., “Performance-BasedExposure Control Limits for Pharmaceutical Active In-gredients,” American Industrial Hygiene Associa-tion Journal, Vol. 57, 1996, pp. 33-42.

12. Harrison, D., “Matching Containment Equipment to theProduct Hazard,” ISPE Conference, Amsterdam, Neth-erlands, December 8-9, 1999.

13. Ryder, M., “Design Requirements Criteria for Contain-ment Device Selection,” ISPE Conference, Amsterdam,Netherlands, December 8-9, 1999.

14. Harrison, D., “Validating the Performance of Engineer-ing Control Systems,” ISPE Conference, Amsterdam,Netherlands, December 5-6, 2001.

15. Koch, M., “Standard Testing of Containment of Connec-tion Systems,” ISPE Conference, Amsterdam, Neth-erlands, December 5-6, 2001.

16. Tahara, S., Watanabe, K., “Construction of Design Proce-dure for Chemical Hazard Facilities. Part 1 of 3,” PharmTech Japan, Vol. 15, No.10, 1999, pp. 1441-1450, inJapanese.

17. Takeda, M., Ito K., Shirokizawa, O., and Watanabe, K.,“Construction of Design Procedure for Chemical HazardFacilities. Part 2 of 3,” Pharm Tech Japan, Vol. 15,No.11, 1999, pp. 1619-1630, in Japanese.

Page 33: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Containment Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 9©Copyright ISPE 2003

18. Masuda, M., Kawai, M., Tozaki, K., and Watanabe, K.,“Construction of Design Procedure for Chemical HazardFacilities. Part 3 of 3,” Pharm Tech Japan, Vol. 15,No.12, 1999, pp. 1769-1780, in Japanese.

19. Takeda, M., “Trends in Handling Potent Compounds inJapan,” ISPE Conference, Amsterdam, Netherlands,December 8-9, 1999. JP2001-222636

20. Suzuki, O., “Design Criteria for Barrier Isolation Systemsand Containment Evaluation in Pharmaceutical Facili-ties,” ISPE Conference, Amsterdam, Netherlands,December 5-6, 2001.

21. Whyte, W., “An Introduction to the Design of Clean andContainment Areas,” Cleanroom Design, ed., W. Whyte,Johm Wiley & Sons, Inc., 1995, pp. 1-22.

22. Cocker, N., “Vale III Isolation Validation - GuaranteedProtection,” ISPE Conference, Amsterdam, Nether-lands, December 8-9, 1999.

23. Pujara, C.P., Kildsig, D.O., “Effect of Industrial ParticleCharacteristics on Airborne Emissions,” Containmentin the Pharmaceutical Industry, Drugs and Phar-maceutical Sciences, Vol. 108, ed., Wood, J.P., MarcelDekker, Inc., 2001, pp. 29-53.

AcknowledgmentsThe authors are deeply indebted to Toshio Sagara, ManagerMamoru Numata, Group Leaders Tetsuo Fujioka and ShinichiYamagami, Senior Managers Makoto Miyamoto, KeiichiroWatanabe, and Shinichi Osada of the Industrial ProjectDivision, JGC for their helpful suggestions and valuablecriticism during the evaluation of the containment facilitiesand the preparation of this article. The authors also wouldlike to thank Michael Lucey and Michiko Ishizuka of Indus-trial Project Division, JGC for their excellent support inimproving this article.

About the AuthorsOsamu Suzuki is a Senior Researcher in TechnologiesResearch Center of JGC Corp and recently joined a researchproject of evaluating containment performances in variouspharmaceutical facilities designed by JGC. He publishednearly 30 articles in the field of biomedical science, surfacechemistry and pharmaceutical engineering including a Phar-maceutical Technology article relating to mechanism study tocause rouge in a pure steam generator. He received a PhDfrom Tohoku University Graduate School of Medicine in1991; was Visiting Scientist at Forsyth Research Institute,Physical Chemistry Department (Boston, MA, USA) from

1992 to 1994, and a Research Associate in Japan FineCeramics Co. Ltd, a subsidiary company of JGC, from 1986 to1994. He has been an ISPE member since 2001.

JGC Corp., Technologies Research Center, 2205 Narita-cho, Oarai-machi, 311-1313 Japan, tel: +81-29-267-0165,email: [email protected].

Morihiko Takeda is a Manager in the Industrial ProjectDivision of JGC Corp and has recently assumed the leader-ship in the containment evaluation research project. Heproposed the design criteria for the pharmaceutical contain-ment facilities described in this article. He was a speaker atthe 1999 ISPE Amsterdam Conference, Containment of Po-tent Compounds. He has been acting as lead engineer tovarious projects for parenteral and solid dosage productsmanufacturing plants for domestic and multinational com-panies. He has a BSc in mechanical engineering from SaitamaUniversity in 1981. He joined JGC in 1981. He has been anISPE member since 1999.

JGC Corp., Industrial Project Div., Yokohama World Op-eration Center, 3-1, Minato Mirai, Nishi-ku, Yokohama 220-6001, Japan, email: [email protected].

Koji Tanaka has been a Research Associate in the Technolo-gies Research Center of JGC Corp since 1999 and has beendedicating himself to obtaining and analyzing the pharma-ceutical containment evaluation data. He has experience inanalyzing various containment systems including nuclearfacilities that are used to keep the reduced condition insidethe glove box. He learned various handling procedures forhazardous chemicals, poisonous gases, and radioactive sub-stances through these containment evaluations. He has anMSc in analytical chemistry from Kyoto University GraduateSchool of Engineering, Department of Molecular Engineer-ing by studying syntheses and properties of organic conduc-tors in 1999. He has been an ISPE member since 2001.

JGC Corp., Technologies Research Center, 2205 Narita-cho, Oarai-machi, 311-1313 Japan, email: [email protected].

Mikio Inoue is a Chief Engineer in the Industrial ProjectDivision of JGC Corp, consulting for research projects includ-ing the containment evaluation research project. He hasexperience in the design of aseptically operating mammal cellculture systems from laboratory to industrial manufacturingscale. He also has experience in GMP relating to pharmaceu-tical designs and is taking part in basic design schemes in thefield of cell processing and tissue engineering manufacturingplants regarding the building layout and zoning from thepoint of view of GMP regulation. He received a PhD fromTohoku University Graduate School of Science in 1975. Hejoined JGC in 1975. He experienced process engineering inmainly chemical industrial fields before joining the healthcareindustrial field. He has been an ISPE member since 2000.

JGC Corp., Industrial Project Div., Yokohama WorldOperation Center, 3-1, Minato Mirai, Nishi-ku, Yokohama220-6001, Japan, email: [email protected].

Page 34: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

Step-Variable Air Volume Fume HoodControl - A Case Study

by Sarla M. Patel, PE and Martin J. Wendel Jr., PE

This articledescribes aninnovativedesign of afume hoodcontrol system.

Introduction

This article describes an innovative de-sign of a fume hood control system. Thisdesign innovation combines (for a spe-cific chemical development laboratory

facility) the best features of traditional con-stant volume and variable volume fume hoodcontrols, while avoiding some of their perceiveddisadvantages.

Design Challenge -Laboratory Fume Hood Control

Fume hoods are critical elements in the designand operation of a pharmaceutical researchand development laboratory. Fume hoods areessential in many applications to protect work-ers from exposure to chemical fumes or otherhazardous substances. At the same time, fumehoods have a significant impact on the labora-tory construction costs and operating costs be-

Figure 1. Bench topfume hood - shown withsashes closed.

cause of the equipment and energy required toexhaust air through the fume hoods, and toprovide conditioned ventilation air to the labo-ratory spaces.

One of the many important laboratory de-sign issues associated with fume hoods is theselection of a fume hood control strategy, whichthen leads to the control strategy for overalllaboratory airflow and pressurization. Con-stant volume fume hood control has been usedin many laboratories for years, and is generallyconsidered to be a solid, reliable approach tosafeguard workers. For laboratories with largenumbers of fume hoods, constant volume fumehood control results in enormous energy useand operating costs since the same amount ofair must be conditioned and exhausted throughthe fume hoods whether they are in use or not(that is, whether the fume hood sashes are openor closed). More recently, variable volume fume

hood controls havebeen developed andinstalled to reduceenergy consumptionand operating costs infume hood-intensivelaboratories by reduc-ing air flow through afume hood when thesash is closed. Manyof these systems,which typically main-tain a constant fumehood face velocity tocapture fumes andprotect laboratoryworkers, also are gen-erally considered to besolid, reliable systemswith good trackrecords.

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 35: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

The technical issues associated with fume hood controlstrategy selection can be quantified and analyzed in terms ofperformance, installed cost, operating, and maintenance costs.However, it is essential that this system selection (and mostothers) includes consideration of owner acceptance, includingthe scientific research community that will use the labs andthe building engineers who will maintain the systems. Thebuilding design must be responsive to the needs of the peoplewho will work in the building. Sometimes, as a result of theconsideration of both human and technical requirements, thebuilding design professional has to think outside the con-straints of pre-packaged solutions to achieve design excel-lence. This article discusses one example of this type ofinnovation, and the benefits of the resulting system design.

The Case StudyThe building design project was a 75,000 gross square foot(6,970 gross square meters) chemical development facility.The scientific work planned for this facility required fumehood-intensive laboratory spaces. A typical 735 square foot(68 square meters) laboratory module was planned to includea total of six fume hoods, four bench top hoods, and two walk-in hoods.

The operating profile of the fume hoods was identified bythe hood users as generally being one or the other of twoscenarios: sashes either 50% open or fully closed. The hoodsashes would be opened 50% either when experiments are“setup” within a hood, or certain operations are performedthat require the user to frequently be in-and-out of the hoods.At all other times, the sashes would be kept closed. Situationswhere the sashes would be partially open at a point some-where between 50% and fully closed were not considered a“real world” condition that would occur in practice.

The building design professionals proposed to design Heat-ing, Ventilating and Air-Conditioning (HVAC) systems ac-counting for diversity in fume hood use to achieve constructionand operation savings. That is, if fume hood exhaust can bereduced when the sashes are closed, the exhaust systems canbe sized for a diversified load since only a fraction of all fume

hood sashes will be open at any one time. Since the high fumehood density in this laboratory meant that the room air changerate was exhaust-driven, the diversity also permitted reduc-tions in supply air conditioning system and chilled watersystem sizing. The fume hood operating diversity was based onthe agreement by the scientific researchers to institute aprogram of fume hood sash management so that the maximumcombination of hoods with sashes open at the same time (on aper-lab basis) would be two bench top hoods and two walk-inhoods, or four bench top hoods and one walk-in hood.

Design CriteriaThe solution to the design challenge had to meet the followingcriteria:

1. The solution must maintain safe conditions for scientificresearchers and other laboratory workers.

2. The solution must satisfy user demands for simplicity ofinstrumentation, controls, and operations.

3. The solution must permit reduction in fume hood exhaustairflow to allow economic HVAC system design based onoperating diversity.

4. The solution must permit further reduction in fume hoodexhaust airflow during operation to lower energy con-sumption and costs without a substantial maintenancecost penalty for ongoing component and system calibra-tion and adjustments.

Traditional Design Alternatives -CAV and VAV Control

The two design alternatives traditionally considered for thistype of laboratory facility are Constant Air Volume (CAV)control, which utilizes CAV boxes controlled to satisfy aconstant airflow setpoint, and Variable Air Volume (VAV)control, which utilizes VAV boxes configured to providevariable system airflow. Since the ultimate design solutionincluded a mix of the concepts involved in these two tradi-tional designs, their application will be briefly reviewed.

Constant Volume Fume Hood (Figure 2)Constant volume bypass hoods (CAV) typically feature hoodsashes that operate in tandem with a “bypass” that is reverseacting with the position of the sashes. This arrangementprovides a constant exhaust flow, measured in cubic feet perminute (cfm), through the fume hood regardless of the sashpositions - Figure 2. The exhaust airflow through the hood ismaintained constant through the use of a CAV box installedin the ductwork serving the hood that is calibrated andcontrolled to always provide a constant air volume CAVexhaust flow setpoint. This CAV box has an airflow measur-ing device and associated controls that modulates an “airvalve” the same way a VAV box is configured. The onlydifference is that with a CAV box, the “control system”modulates the CAV box to maintain a programmed constantFigure 2. Constant volume bench top fume hood.

Page 36: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

flow, regardless of “upstream” system dynamics in airflowand pressures. If we look at a typical CAV bench top fumehood, when the hood sashes are fully open, the bypass is fullyclosed, and the air flows through the sash opening (andairfoil). When the sashes are fully closed, the same quantityof air flows through the bypass (and airfoil). The CAV boxesare applied to the system to draw a constant exhaust airflowrate from the hood regardless of whether the exhaust airflows through the sash opening or the bypass.

CAV hoods are high energy users because there is noreduction in exhaust air out of the hoods when the sashes arepartially or fully closed. No reduction in exhaust air alsomeans no reduction in conditioned supply (makeup) airdelivered to the lab to balance the exhaust air from the hood.

One energy savings alternative typically applied to CAVhoods is to index (or “set back”) the hoods to a lower exhaustflow when the laboratories are unoccupied. During this unoc-cupied mode, the exhaust air through the hood is reduced tosome percentage of full flow (60% for example), which alsoresults in a direct reduction of supply air to the space wherethe hood is installed. This flow set back approach has typi-cally been applied on a lab-by-lab basis and sometimesrequires additional construction expense to provide an occu-pied/unoccupied indication outside of the laboratory.

One or a combination of the following methods may controlthe occupied/unoccupied mode changeover:

1. globally for all laboratories via a time schedule

2. manually via a hand switch provided for each hood orgroup of hoods

3. manually via a relay wire to the light switch in the spacewhere the hood is installed to index the hoods to theoccupied mode when the lights are turned on and theunoccupied mode when the lights are turned off.

4. automatically via motion sensor technology detecting thepresence of a researcher

5. automatically via monitoring the position of the sashes todetect sash closure - unoccupied when hood sash is fullyclosed and occupied when hood sash is not fully closed

Variable Air Volume (VAV) HoodsIn VAV fume hoods, the airflow through the hood variesdepending on hood sash position or hood differential pres-sure. Figure 3 depicts airflow with hood sashes open andclosed respectively. In VAV hoods, the exhaust airflow throughthe hood is typically varied to maintain a constant facevelocity through the hood sash opening through the use of aVAV box installed in the ductwork serving the hood. Thesupply and exhaust airflow is controlled by sensing the hoodsash position or by sensing airflow via an anemometer lo-cated in the hood side wall.

Hood sash position control utilizes a potentiometer orsimilar position-sensing device attached to the sash througha cable. When the hood sash position changes, the sensingelement responds to the change in sash position. The associ-ated controller feeds back to the Building Management Sys-tem (BMS) to determine the required air quantity and controlof the exhaust air regulator to achieve a position (and corre-sponding exhaust airflow) that corresponds to the new sashposition.

Hood side wall anemometer control utilizes an airflowsensor called a thermal anemometer to infer hood face veloc-ity. The sensor measures air velocity passing through a holein a side wall of the hood, which has been shown experimen-tally to represent an average face velocity. When the hoodsash is opened, the sensor measures the reduction in facevelocity and sends an appropriate signal to the exhaust airregulator, via the BMS, to increase the airflow until the facevelocity setpoint is reached. When the hood sash is lowered,the sensor measures a velocity increase and sends a signal todecrease airflow.

Comparing Traditional CAVand VAV Fume Hood Control

Comparing traditional CAV and VAV fume hood control, onemay observe that CAV control is free of complex instrumen-tation and hence inherently more reliable. The hood operat-

Figure 3. Variable volume bench top fume hood.

“Sometimes, as a result of the consideration of both human and technicalrequirements, the building design professional has to think outside the constraints of

pre-packaged solutions to achieve design excellence.”

Page 37: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

ing integrity is maintained because the required amount ofair is available all the time. The primary disadvantages ofCAV control are the energy cost of exhausting unused condi-tioned air when the sashes are closed, and high air handlingunit and ductwork costs since no diversity can be taken withCAV.

Traditional VAV control provides more economical opera-tion when the sashes are less than fully open as it takes onlythe amount of conditioned air required to maintain a mini-mum face velocity with minimal bypass. Constant face veloc-ity is maintained and continuous monitoring and alarming offace velocity is provided. However, there is an additional costof instrumentation for sensors and controllers. Furthermore,VAV control sometimes requires a separate proprietary fumehood control system, independent of (but usually interfacedto) the BMS. This increases installed cost and operating/maintenance costs for the system.

The Design SolutionThe design solution, referred to as Step-VAV control, is acombination of the best features of traditional CAV and VAVcontrols, while avoiding some of their perceived disadvan-tages.

The design solution utilized the fume hood types preferredby the researchers - Figure 1. The hoods were bypass CAVwith taller than standard height hood sashes. Each hood wasprovided with four horizontal sliding sashes in a double trackarrangement so that no more than 50% of the hood sash areacould ever be open at any one time. Whenever a hood sash isopened, its bypass door is closed in the same proportion. Eachsash door is furnished with a door switch. The door switchcontact closes when the hood sashes are fully closed. Thecontact opens when any sash is moved from the fully closedposition. As noted, there are four doors at each hood. Since thecontrol strategy is based on detecting when all the sashes arefully closed, the position of individual sashes is not importantand the fume hood manufacturer wired all switches to pro-vide one common contact closure per fume hood. Each hood isprovided with a low flow alarm light, supplied by the hoodmanufacturer. The low flow alarm and auxiliary alarm con-tacts are activated whenever hood face velocity drops belowthe set flow. Hood airflow cfm (cubic feet per minute) require-ments are indicated in Table A.

The laboratory exhaust system is comprised of constantvolume two-position air terminal boxes, one per hood. Theexhaust is ducted from each hood directly to its dedicatedexhaust box. The laboratory supply system is comprised oftwo VAV supply boxes that serve each lab module. There is areheat coil down stream of each VAV box. Air is delivered tothe room through diffusers.

Lab general exhaust was not required as the laboratory

module was a “hood driven” space. This means that evenwhen all of the hoods were closed, the minimum combinedairflow setting for all hoods with all sashes closed was a valuethat required “make-up” supply air flow from the air handlingsystem that was beyond that required to offset the heat gainsin the laboratories.

The hood setback exhaust airflow quantity was found,through factory smoke testing, to provide good mixing condi-tions in the hood with air flowing through the bypass in thelow flow condition.

Cost ComparisonThe first cost for a constant volume hood control installationis significantly less than that of a variable air volume hoodcontrol installation for the same size hood. Table B is asummary of the major hardware elements along with “aver-age” associated costs that comprise a typical CV and VAVcontrol system installation for a lab module featuring twofume hoods. The HVAC system serving the model profiledhere has a dedicated “exhaust box” for each hood, and a singlesupply air box that provides makeup air for the hoods andconditioning for the space.

Building Management Systemand Instrumentation Requirements

The microprocessor-based Direct Digital Control (DDC) Build-ing Management System (BMS) monitors and controls thelab/hood air flow and space temperature. This system is anextension of the BMS used for total building HVAC systemcontrol and monitoring. This Step-VAV control design doesnot require a proprietary lab airflow control system.

Instrumentation requirements for laboratories using Step-VAV control are very simple compared to labs using tradi-tional VAV control. Each exhaust box utilizes the box supplier’sstandard pneumatically operated two-position control prod-uct. When pneumatic control signal pressure is 0 psig (101kPa), the box is at maximum exhaust CFM, and whenpneumatic control signal pressure is 13 psig (224 kPa), thebox is at minimum exhaust CFM. The pneumatic signal isprovided by the BMS, 13 psig (224 kPa) when all sashes at the

Table B. CAV/VAV hood control installation cost comparison.

Constant Volume Hood Control Installation

Terminal exhaust box controller $2,000

Terminal supply box controller $2,000

Hood alarm $1,000

Unoccupied/occupied override switch $500

Total $5,500

Variable Air Volume Hood Control Installation

Fume hood controller with sash sensor, hood alarm panel $8,000and exhaust box control

Lab control panel with flow tracking, supply box and $7,000room temperature control

Total $15,000

Table A. Fume hood exhaust volumes.

Hood Type Sashes 50% Open All Sashes Fully Closed

Bench Top 1140 CFM (538 L/S) 475 CFM (224 L/S)

Walk-In 2220 CFM (1048 L/S) 890 CFM (420 L/S)

Page 38: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

hood are fully closed, and 0 psig (101 kPa) when one or moresashes is not fully closed.

Control response time of the instrumentation used toindex the setback was found to be fast enough to maintainhood containment while switching the exhaust boxes be-tween modes.

Supply boxes are controlled by the BMS supplier’s stan-dard product called a “Remote Control Panel (RCP).” Thispanel is a stand-alone controller and communicates on a“peer-to-peer” basis with other RCPs over a dedicated com-munication network. For reliability, one RCP was providedper lab. In the event of failure of a RCP, only one lab would beaffected. Because the RCPs have a modular design and weresized to provide required input/output points, the cost pen-

alty to provide an individual RCP per lab (compared tocontrolling several labs from one RCP) was not prohibitive.Each supply box included an airflow probe (furnished by thebox suppler), a differential pressure-type airflow transmit-ter, a damper (furnished by the box supplier), and an electricdamper actuator.

The reheat coil valve actuator is electrically operated.Space temperatures are measured by wall mounted tempera-ture sensor/transmitters. There are also two duct mountedtemperature sensors/transmitters, one in each supply duct.The reheat coil valves are controlled using space temperaturesensors. The duct mounted temperature sensors are used foranticipatory BTU control of the reheat control coil valves –Figure 4.

Figure 4. Typical Step - VAV Lab Module Controls.

Page 39: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Control Sequence of Operations1. The BMS monitors the hood sash position for each fume

hood via sash end switch contact.

2. The BMS changes each exhaust box CFM setpoint basedon its sash position.

3. The BMS calculates total exhaust CFM by summing airexhausted from each hood with sash open and each hoodwith sash closed. Using the result, the DDC Systemcalculates required supply air CFM (supply CFM = ex-haust CFM – constant transfer CFM). As there are twosupply boxes serving each lab module, each box willprovide half of the total required CFM. The BMS modu-lates the supply box dampers to maintain the supply CFMsetpoint.

4. The BMS limits supply CFM to the maximum design limit- to allow a maximum of two bench top hoods and two walk-in hoods open at the same time, or four bench top hoods andone walk-in hood. If a researcher opens sashes in morethan the design allowance of fume hoods, the BMS acti-vates a strobe light in the lab indicating “TOO MANYHOODS OPEN – CLOSE HOOD SASHES.”

5. If hood sashes are not closed immediately upon activationof the strobe light, exhaust air from each hood reduces,resulting in a further drop in face velocity. When the facevelocity drops to its preset low limit, a “LOW FACEVELOCITY” alarm light at each hood (provided by hoodmanufacturer) is activated.

6. The BMS maintains space temperature setpoint by modu-lating reheat coil control valves from the signal of onespace temperature sensor and two duct mounted tempera-ture sensors, one in each discharge duct in cascade mode.

Operating Reduction in AirflowThe system is designed to accommodate a maximum of twobench top hoods and two walk-in hoods, or four bench tophoods and one walk-in hood in simultaneous operation withsashes 50% open. Since the hood sashes are typically openedfor experimental setup and closed otherwise, the Step -VAVdesign permits significant flow reduction within each lab.The required lab air flows are the sum of the flows for hoodswith sashes 50% open plus flows for the hoods with all sashesclosed. For example, with one walk-in hood open, the lab airflow for two walk-in hoods is 1 x 2,220 cfm (1048 L/s) + 1 x 890cfm (420 L/s) = 3,110 cfm (1468 L/s). The different hoodoperating conditions and corresponding airflow volumes areindicated in Table C.

As can be seen by inspection of the values in Table C, StepVAV control for the typical labs in this case study providesstepped modulation of air flow from a maximum of 7,670 cfm(3,620 L/s) to a minimum of 3,680 cfm (1,737 L/s) in 665-cfm(314-L/s) increments. In this application, Step VAV controlprovides the same operating cost benefits as a traditionalVAV control approach. Since the researchers typically opentheir hoods for experimental setup and keep them closed atother times, the lab airflow demand is stepped by its natureand any additional airflow reductions provided by traditionalVAV control would be only transitory.

System BenefitsThe design solution implemented for this project providessignificant benefits to the building owner, including thefollowing:

1. This design does not require the complex instrumentationand control of conventional VAV control systems, such asa VAV exhaust box controlled from hood sash position andtracking supply with each change in each hood sashposition. Monitoring individual hood sash positions for the

Table C. Hood sash/lab air flow combinations.

Number of Walk-In Hoods with Number of Bench Top Hoods with Laboratory Air Flow VolumeSashes 50% Open <airflow> Sashes 50% Open <airflow> for this Configuration

2 <total 4,440 cfm/2.096 L/s> 2 <total 3,230 cfm/1,525 L/s> 7,670 cfm (3,620 L/s)

2 <total 4,440 cfm/2.096 L/s> 1 <total 2,565 cfm/1,211 L/s> 7,005 cfm (3,306 L/s)

2 <total 4,440 cfm/2.096 L/s> 0 <total 1,900 cfm/897 L/s> 6,340 cfm (2,992 L/s)

1 <total 3,110 cfm/1468 L/s> 4 <total 4,560 cfm/2,152 L/s> 7,670 cfm (3,620 L/s)

1 <total 3,110 cfm/1468 L/s> 3 <total 3,895 cfm/1,838 L/s> 7,005 cfm (3,306 L/s)

1 <total 3,110 cfm/1468 L/s> 2 <total 3,230 cfm/1,525 L/s> 6,340 cfm (2,992 L/s)

1 <total 3,110 cfm/1468 L/s> 1 <total 2,565 cfm/1,211 L/s> 5,675 cfm (2,679 L/s)

1 <total 3,110 cfm/1468 L/s> 0 <total 1,900 cfm/897 L/s> 5,010 cfm (2,365 L/s)

0 <total 1,780 cfm/840 L/s> 4 <total 4,560 cfm/2,152 L/s> 6,340 cfm (2,992 L/s)

0 <total 1,780 cfm/840 L/s> 3 <total 3,895 cfm/1,838 L/s> 5,675 cfm (2,679 L/s)

0 <total 1,780 cfm/840 L/s> 2 <total 3,230 cfm/1,525 L/s> 5,010 cfm (2,365 L/s)

0 <total 1,780 cfm/840 L/s> 1 <total 2,565 cfm/1,211 L/s> 4,345 cfm (2,052 L/s)

0 <total 1,780 cfm/840 L/s> 0 <total 1,900 cfm/897 L/s> 3,680 cfm (1,737 L/s)

Page 40: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Case Study - Fume Hood Control

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

horizontal sash arrangement with four doors is not re-quired.

2. The first cost of Step VAV is significantly less than the firstcost of traditional VAV for the same fume hood applica-tion.

3. There should be no differential cost to the Owner in theprogramming of Step VAV versus traditional VAV as theBMS system would have to be programmed regardless ofthe HVAC operating scheme.

4. This design is more robust than traditional VAV controlbecause it is more tolerant of variations in component(such as exhaust damper) response times. Rather thantrying to “chase” traveling sashes, this design provides fullexhaust flow as soon as any sash is moved from the fullyclosed position, and maintains full exhaust flow until allsashes are fully closed.

5. Airflow control is provided by the time proven technologyof the BMS (DDC System), which is simple, cost effective,and reliable, without introducing additionalmanufacturer’s systems (that could add complexity andcost to building maintenance) into the facility.

6. It is anticipated that overall maintenance costs (typicallyaveraging approximately $100/per sensor) will be less forStep VAV as there are fewer BMS points associated withthis scheme over traditional VAV applications.

7. In this application, where the hood sashes will generallybe opened only while setting up experiments within a hoodand sashes will be kept closed at all other times, step VAVcontrol is more appropriate to the owner’s intended opera-tion compared to conventional VAV. Capital cost savingsand operating/energy cost savings are achieved throughthe simple addition of a single point, “go/no-go” measure-ment at each fume hood. Put another way, if a traditionalVAV system were designed into the case study facility, itwould be used in a manner that profiles the way a “Step-VAV” system would be used, so the operating costs wouldbe similar. The key is, why pay the high first cost oftraditional VAV if you are not going to use the system thatway? The true savings are lower first cost with “Step-VAV”control over traditional VAV control.

SummaryThe implemented hood control design is Step-VAV providingonly the required amount of air to each hood. This minimizesthe capital cost (through diversity in equipment sizing) andoperating costs. The control system is simple, reliable, andeasy to maintain. It utilizes the researchers’ preferred con-stant volume bypass hoods. The only automatic sensingrequires hood sash end switch position monitoring by theBMS, using simple proven technology. Exhaust flow for each

hood is controlled in two steps; maximum flow when any sashis open, and minimum flow when all hood sashes are closed.In the present application of six hoods, there are 13 steppedflow conditions, providing a stepped approximation of VAVcontrol. Supply flow is controlled by the BMS to maintaintransfer airflow directionality. The installed cost of thissystem is lower than traditional VAV and the resultingdesign is very flexible.

About the AuthorsSarla M. Patel, PE is a Senior Project Engi-neer at Kling. Patel has more than 35 yearsof experience in instrumentation and con-trol, electrical engineering, reliability, main-tainability, and computer microprocessor ap-plication. In her present position, she is re-sponsible for project engineering, design anddevelopment, and for equipment selection.

Patel’s experience includes systems for automatic tempera-ture control, building automation, energy monitoring andcontrol, security, fire alarm and smoke detection controlsystems, motor control centers, sound and communications,lighting, data communications, telephone electronics, pro-cess control, and local area networks. She has provided theengineering services for Glaxo Inc.’s Pharmaceutical Head-quarters Campus in Research Triangle Park, NC, WyethResearch Inc.’s Great Valley Corporate Center in Frazer, PA.,and multiple research laboratory renovations for Merck andCo. Patel is a member of the Instrumentation Society ofAmerica and the Institute of Industrial Engineers. She holdsa bachelor’s degree in electrical engineering from the Univer-sity of Michigan and a master’s in industrial engineeringfrom the University of Pittsburgh. She can be contacted at:[email protected].

Martin J. Wendel, Jr., PE is an Engineer-ing Design Principal at Kling. Wendel isresponsible for engineering leadership andoversight of all mechanical services disci-plines, process plumbing, fire protection, andinstrumentation and controls. Wendel hasexpertise on a wide variety of office, researchand development, and computer facilities,

having designed mechanical systems for high-technologylaboratories, pharmaceutical development facilities, US Fed-eral Government buildings, and manufacturing facilities. Hehas provided professional services for Merck and Co. inWilson, NC, Glaxo Inc.’s Pharmaceutical Headquarters Cam-pus in Research Triangle Park, NC, and Wyeth Facilities inPearl River, NY. Wendel is a member of the American Societyof Heating, Refrigeration and Air Conditioning Engineers,and ISPE. He holds a bachelor’s degree in mechanical engi-neering from Drexel University. He can be contacted at:[email protected].

Kling, 2301 Chestnut St., Philadelphia, PA 19103, (215)569-2900.

Page 41: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

An Automation and ValidationStrategy for an Automated APIManufacturing Plant

by Jorge Manuel C. Pastilha

Redefining anAutomation andValidationStrategyallowed a plantto be revisedand designed fora new treatmentproduct withinthree months ofstart-up.

Introduction

This article describes the design, con-struction, commissioning, and opera-tion of a 200 m3 (7063 ft3) automatedproduction plant for APIs meeting US

FDA cGMP at Hovione, FarmaCiencia SA, Por-tugal.

The construction of a new automated plantwas a great challenge that led the company toredefine its Automation and Validation Strat-egy. This new strategy allowed a plant, whichhad been designed to produce a single product(X-Ray contrast media), to be revised and de-signed for a new HIV treatment product withinthree months of start-up. In February 1998, thethree consecutive validation batches were con-cluded. Three months later, the 25-day processwas stable, delivering a new batch every 72

hours and a successful FDA pre-approval in-spection was complete.

An increasing demand for safety and qualityrequire that these factors are embedded in themanufacturing process, and therefore, in theproduct. Automation of manufacturing pro-cesses, when implemented under quality rules,has proven to be an important factor both inincreasing safety and quality of the environ-ment and the efficiency of the manufacturingprocess itself.

Initial Design“The biggest problem found in most projects is alack of clear and complete scope definition.1”Based on the concept that the initial project stepsare critical for success of a project, the companyincreased the effort during the design phase.

Figure 1. Parallel tasksduring Validation of anAutomated API Plant.

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 42: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 2. Overview of the Automation Strategy.

Automation StrategyThe automation strategy focused on modularity which pro-vided a simple solution for the complexities involved in thedesign, construction, commissioning, validation, and renova-tion of the automated API plant.

The modularity concept was applied in three distinctareas:

1. Equipment, Materials, Instruments and Spare Parts(Project and Construction)

2. Functionalities (Automation)

3. Documentation and Tests (Validation)

Modularity meant:

• proven technology and reliable equipment• reduced number of different materials• instrumentation should be nearly identical• devices (valves, pumps and motors) must be normalized• functionality (heating, cooling, temperature control, pres-

surization, depressurization, vent, charge materials, dis-charge, etc.) should be as generic and as versatile aspossible, while, at the same time, simple

• the Project Team must agree on standard formats forspecifications and testing documentation.

Advantages of the modularity concept:

1. guaranteed uniformity along the time between projects2. meeting of the quality requirements3. reduction in the implementation, testing, and validation4. reduction in the number and cost of spare parts5. reduction in the time required for training of operation

and maintenance personnel

Example Application of the Modularity ConceptA reactor is composed of a vessel, an agitator, discharge andthermal fluid pumps, on-off valves, control valves, condens-ers, piping, and field instrumentation. Reactors can havedifferent volumes, be constructed of different materials, andhave different quantities of valves or associated piping. Suchvariations may cause individual reactors to be consideredunique.

The pumps, valves, and motors that compose the reactorare considered generically as devices. The communicationbetween the Distributed Control System (DCS) and thedevices is achieved through electrical signals, known asInputs/Outputs (I/Os).

The devices may be grouped according to their functional-ity on the reactor (such as adjusting temperature, controllingpressure, and charging material in the reactor). These func-tional groups of devices are designated as Equipment Mod-ules (EQMs).

Each equipment module performs a different action, suchas heat, cool, or maintain temperature; pressurize, depres-

Definition of User Requirement SpecificationsA key document, “Process Book/Basic Engineering,” wasapproved almost a year after the first steps to clarify therationale of the project and its location were taken. Thisdocument described in concise terms the X-Ray contrastmedia production process, and presented the main equip-ment to be installed with specifications, which togetherconstituted the User Requirement Specifications.

The dimensions of the project clearly indicated that acompletely new approach to validation was necessary:

• 21 reactors, 6 centrifuges, 3 dryers, more than 20 tanks,liquid film evaporator, reverse osmosis systems, a produc-tion and distribution purified water system, cleanrooms,etc.

The introduction of a high level of automation (the intentionwas to operate the installation from a central control room)also supported the need for a new approach.

Definition of Responsibilitiesand Creation of a Dedicated TeamThe project required a multidisciplinary team, but definingthe precise membership of the team proved one of the mostdifficult decisions of this initial phase. Ultimately, a team ofhighly motivated engineers with experience in several disci-plines was created in conjunction with guarantees of ‘top-level’ involvement. The following three sub-teams were thenidentified:

1. Project and Construction2. Automation3. Validation

Planning Activities -The Importance of Parallel PathsDue to the high level of automation, it became clear thatautomation and validation tasks (Figure 1) needed to beexecuted concurrently with the construction and commis-sioning of the facility. It was equally important that thetiming of these tasks was communicated effectively to allmembers of the team.

Automation and Validation StrategyKnowing the timeline and the resources involved provedessential and the project team decided to make the project anopportunity to develop an Automation and Validation Strat-egy.

Page 43: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

surize, or maintain pressure, charge, discharge, or agitate.Each action causes the DCS to act on the devices to achievethe expected result (Setpoint) for a specific variable (e.g.,temperature, pressure, or level) measured by a field instru-ment. Each action will be executed in a specified way thatconstitutes a ‘control strategy.’ To ensure both equipmentand personnel safety, interlocks are created at the equipmentmodule level.

A reactor is generally designated as a process unit, whichgroups the equipment modules with the different controlstrategies and provides all necessary interfaces between thecontrol system and the operators.

The strategy followed for a reactor can be applied to otherprocess units, such as tanks, dryers, or centrifuges. Figure 2shows an overview of the automation strategy. A plant isregarded as a group of process units, each consisting ofequipment modules that are functional groups of fieldinstruments, and devices that communicate with the fieldinstruments through the I/Os.

Each box shown in Figure 2 corresponds to a softwaremodule that must be fully documented, developed, and tested.Once validated, a module is approved and released, may beduplicated, or cloned, in various combinations, providinggreat utility and flexibility. For example, if a plant has onethousand ‘fail close on-off valves with closed limit switch,’then one generic valve will be documented, developed, tested,and approved. This will be ‘cloned’ one thousand times,assuring that all valves have the same behavior.

Validation StrategyThe project development phases were performed in parallelto, and partially overlapping with, the validation activityphases of Design Qualification (DQ), Installation Qualifica-tion (IQ), Operational Qualification (OQ), and PerformanceQualification (PQ).

The aim of the company was to develop a strategy thatallowed automation and validation of new API plants with-out repetition of much of the work.

Figure 3 shows the steps, support documentation, andvalidation phases for the implementation of an automatedsystem. Four phases in the implementation of the automatedsystem were recognized:

• Phase 1 - Preparation and release of Generic Tools

• Phase 2 - Application and customization of Generic Toolsfor a specific project; installation and connection of systemhardware, testing of functionality

“Once validated, a module is approved and released,may be duplicated or cloned, in various combinations,

providing great utility and flexibility.”

Table A. Building 15-key dates.

Civil works Sep 1995 - Feb 1997

Mechanical construction Nov 96 - Aug 97

Instrumentation installation Feb 97 - Oct 97

Automation Project definitions and Quality plan Sep 96 - Mar 97

Automation “Generic Tools” development and Nov 96 - Oct 97implementation

Graphics, Functional and Design Specs, Database Apr 97 - Jul 97Generation and Implementation for X-Ray product

Re-engineering for the new product Sep 97 - Nov 97

FDA Pre-Approval Inspection Mar 98

500 kg batches, one every 72 hours Jun 98

500 kg batches, one every 48 hours May 99

• Phase 3 - Validation of the system for the production of aspecific product

• Phase 4 - Production support and system maintenance

Phase 1: Preparationand Release of Generic ToolsAll functionality that may be applicable in more than oneproject, had to be defined, programmed, and tested duringPhase 1.

Examples of functionality that were considered “GenericTools” and which needed to be developed during Phase 1,included:

• functioning of valves, motors, or pumps• grouping of valves by function• methods of cooling or heating equipment• rules for user access• alarming, trending, and reporting policy• security procedures (backup and restore)• recording of the audit trail

Step 1 consisted of defining documentation for generic systemfunctionality (Generic Tools). Step 2 consisted of designingand implementing the Generic Tools, which were documentedin the Design Specifications. Step 3 consisted of testing thefunctionality defined in Step 1, following the proceduresgiven in the Generic Test Procedures. After functionality wasassured (all tests had passed), the corresponding GenericTools were considered released, and therefore, ready to use inthe next phase.

Page 44: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 3. Implementation Phases for an Automated System.

Phase 2: Application and customization ofGeneric Tools for a specific project; installationand connection of system hardware, testing offunctionality

This phase consisted of developing the automated system fora specific project, using the released Generic Tools, comple-mented with specific documentation.

Each project required specific documentation:

Page 45: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

• Validation Plan: detailing the validation activities to becarried out and the list of tests that were applicable to theproject

• Implementation Specification: defining which GenericTools were applicable and corresponding customization

If a Generic Tool did not deal with a situation, Step 1 of Phase1 was initiated to produce and release a new Generic Tool orrevise an existing one, according to a defined Change Controlprocedure. Completion of customization concluded the De-sign Qualification.

Hardware installation, field connection, and correspond-ing tests composed the Installation Qualification.

Finally, system functionality for this specific project hadto be tested. After functionality is assured (all tests havepassed), the Operational Qualification is completed (ready tostart production).

Phase 3: Validation of the systemfor the production of a specific productThe third Phase consisted of verifying the system capabilityto execute one specific product, through the production of aset of batches. This Phase occurs each time a new product isto be produced.

Whenever the adaptation of the installation or automatedsystem was necessary to meet the process requirements,Phase 2 had to be initiated according to a Change Controlprocedure.

When the set of batches was successfully completed (indi-cating that the installation and automated system fit theprocess needs), Performance Qualification was consideredcomplete.

Phase 4:This Phase refers to regular validated process production andconsisted of support of production activities and systemmaintenance.

Design Qualification (DQ)This means ensuring that the plant has been designed inaccordance with the Guidelines, as specified, and documented

Figure 4. Productivity: Process Control Learning Curve andTraining.

evidence exists to demonstrate this.The sequence of activities during this stage included:

a) Piping and Instrumentation Diagrams (P&IDs); all pipingdesign documents are validated through the signature ofqualified technical personnel and changes can only bedone through a Change Control system.

b) All purchases are validated through the signature ofqualified personnel authorized for technical aspects. In-struments and electrical components are defined on in-strument list and purchase takes place based on theapproved technical data sheets from the supplier.

c) Visits to suppliers before purchase and tests done bysuppliers are also part of the design validation process.

d) Safety aspects also are to be validated under the respon-sibility of a specific working team. HAZOP will allow thedefinition of efficient operating procedures, reducing theunderlying risks to the process and people. Once the mainrisks are identified, the possible minimization and controlalternatives are studied, and the project is adjusted ifnecessary, to ensure the accomplishment of the applicablesafety and environmental protection limits.

At this validation stage, all Functional Specifications (Phase1), corresponding Design Specifications (Phase 1), and Imple-mentation Specifications (Phase 2) are prepared and ap-proved.

Installation Qualification (IQ)Examples of activities that were carried out during this phaseincluded confirmation of the system layout, mechanical in-tegrity, and wiring continuity.

Operational Qualification (OQ)For each specific project, all the applicable functionality hadto be defined in the Implementation Specifications and all thecorresponding tests had to be listed in the Validation Plan.

Testing of the equipment operability (such as heatingcapabilities of a reactor) was carried out in parallel withqualification of the related control system.

Performance Qualification (PQ)Performance acceptance of the automated system was part ofa Performance Qualification Report for a specific product.

Use of GAMP and 21 CFR Part 11The validation methodology followed was based on the GoodAutomated Manufacturing Practice (GAMP) Guide2 and 21CFR Part 113 and it is application to all Phases of theautomated system implementation – Figure 3. These docu-ments helped to develop a high-quality documentation sys-tem.

The documentation was used to verify and provide evi-dence that the defined project execution was followed andthat the implementation had been documented and testedagainst the functional requirements. All validation activities

Page 46: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 5. Reducing Deviations.

were conducted according to written procedures.At the end of the validation, a “Validation Report of

Manufacturing Facility” summarized all the validation workthat had been performed.

Advantages of Starting Trainingin the Early Stages of the Project

From the early stages of the project, the company ensuredthat the people assigned for the design, construction, instal-lation, and validation activities were fully qualified andtrained to assume such responsibilities.

To ensure the success of the project, and its benefits, asuccessful production team had to be provided. The companyproduction teams were accustomed to manual installationsand to adapting these to an automated plant. All productionoperators were intensively trained, and participated in thesystem validation during the Operational Qualification. Theadvantages were obvious: the production teams became fa-miliarized with the automated system, the equipment, andwith the techniques of dealing with an automated plant.

Change ControlModifications could significantly change performance capa-bility and in some cases, they could completely alter thecharacteristics of a process. Therefore, a change controlsystem had to be in place.

This required a formal monitoring system by which quali-fied representatives from appropriate disciplines could re-view proposed changes that might alter validated status andtake preventive or corrective actions to ensure that thevalidated state of a system would be maintained.

Case Study DetailsIn 1995, the plant was initially and specifically designed toproduce a single product. A decision to produce a new differ-ent product was made in September 1997. The challenge wasto adapt the building, known as Building 15, before the endof October 1997. The building had a surface area of 4500 m²,composed of manufacturing areas (production, utilities, con-trol room, I/O rooms, and motor control centers) and support-ing areas (offices, laboratories, dressing rooms, and socialarea). Table A presents the most important dates, includingthe design, implementation, validation, and productionphases.The decision to have a highly automated plant, had a highcost at different levels:

• investment was about 8% of total plant value

• an excessive number of personnel at every phase exceptoperation

• increased space required {350 m2 (3767 ft2) for system andelectrical cabinets and 50 m2 (538 ft2) for control room}

• engineering and technical documentation required con-siderable care with methodology and evidence of compli-ance with procedures

A large number of documents were prepared during thebuilding’s design, including:

• 247 wiring diagrams

• 2000 loop diagrams

• 320 functional specifications

• 290 design specifications

• 184 test procedures

Validation of an automated plant is more time consumingthan that of a manual plant. The validation of Building 15took 12 months and required more manpower and morequalified personnel. Qualified personnel from Instrumenta-tion, Production, Maintenance, and Quality performed thevalidation 24 hours a day.

All operators working in the building had been operatorsin manual plants. There was no resistance to change whenmoving to an automated plant although intensive trainingwas necessary and involved 2,000 hours of training, averag-ing 60 hours of training per operator.

Conclusion - The Success of theAutomation and Validation Strategy

The record has shown that the decision to have a highlyautomated plant proved to be the correct one. Re-engineeringof the building for a new product was possible in the shorttime period because of the key options taken several yearsbefore, namely the Automation and Validation Strategy.

There are tangible and intangible gains that can be di-rectly related to the high level of automation and the valida-tion strategy followed. The most important tangible gainsare:

Page 47: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Automation/Validation Strategy

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

a) Higher productivity - Building 15 has a productivity levelfour times higher than comparable manual installations.Figure 4 shows that productivity (measured in kg ofproduct per man-hour) improves with time.

b) Reduced number of deviations. Deviations due to humanerror tend to zero very quickly, as shown in Figure 5. Adeviation always carries a high cost, due to the number ofactivities and the number of people involved in:

• Description

• Investigation

• Corrective action

• Follow-up

• Closure

Important intangible gains to consider include:

• reproducibility and liability• yields vary less and improve with time• process variables and actions can be continuously regis-

tered• process data is very easy to consult and compare, which

allows a rapid learning process• ensure best-practices are adopted• operations are safer and in case of an accident the actions

can be taken automatically

Emphasis must be placed on developing the correct Automa-tion and Validation Strategy. This provides a powerful andsystematic “tool” that reduces the efforts of implementing orre-engineering an API plant by reusing fully qualified soft-ware modules and related documentation.

References1. Harding, Jeffrey S., “A Crash Course In Project Engineer-

ing,” Chemical Engineering, July 1998, pp.118-126.

2. GAMP Guide for Validation of Automated Systems inPharmaceutical Manufacture, Version 3.0 (Good Auto-mated Manufacturing Practice Forum, March 1998).

3. Code of Federal Regulations, Food and Drugs, Title 21,Part 11, “Electronic Records, Electronic Signatures” (U.S.Government Printing Office, Washington D.C.).

About the AuthorJorge Manuel C. Pastilha is Director ofLogistics and Control Group at HovioneFarmaCiencia SA in Loures, Portugal. He isresponsible for the development, implemen-tation, validation, and maintenance of auto-mated systems. He has been involved inautomation projects throughout the last sixyears.

Hovione FarmaCiencia SA, Sete Casas 2674-506 Loures,Portugal. Tel: +351 21 982 9000. Fax: +351 21 982 [email protected]; www.hovione.com.

Page 48: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

Practical CIP System Designby David Greene

This articleprovides anoverview of CIPas well as abasis forestablishing theflow rate forequipmentcleaning.

Introduction“Rules of thumb” are often used to deter-

mine the flow required for cleaningequipment and pipelines in thebiopharmaceutical industry. These

“rules” may conflict when the desire to main-tain supply and return line velocities results inaccumulating liquid in the equipment beingcleaned. This article provides an overview ofCIP as well as a basis for establishing the flowrate for equipment cleaning.

Purpose of CIPClean-In-Place (CIP) is the term used in thebiopharmaceutical industry to refer to the pro-cess of cleaning process systems and equip-ment without major disassembly of compo-nents. In addition to cleaning, CIP also assistsin Steam-In-Place (SIP) operations by remov-ing chlorides and proteins. Leaving chloridescould cause stress corrosion when heated andresidual proteins would be “baked” on to sur-faces when denatured by steam.

CIP involves a specific combination of pre-determined manual and automated operationsto perform the cleaning, monitor the opera-tions, and document the results. The state-of-the-art involves a series of dedicated CIP skids,each assigned to a particular process area orfunction, with its own piping system. Figure 1shows a typical, but simplified, CIP systemwith the appropriate instrumentation andvalves.

A properly designed CIP system will use theminimum amount of water, chemicals, andutilities, and produce the minimum amount ofeffluent. It will improve safety and reduce main-tenance by removing human error and providedocumentation for validation by ensuring re-producibility with minimal operator interven-tion. It will maintain product quality and re-duce turn-around time between batches.

TimeTypical biopharmaceutical production opera-tions are based on operating 24 hours per day

seven days a week, commonly referred to as 24/7. CIP is only part of a typical 8-hour turn-around (from dirty to clean) cycle, which alsoincludes SIP and the associated heatup, cooldown, and integrity testing. The majority of thetime is for heating and cooling before and afterSIP, but the time for CIP is important in deter-mining the number of CIP systems required.

Factory Acceptance Tests (FATs) typicallytest the effectiveness of spray balls through theuse of a Riboflavin coverage test. This testensures that the vessel internals are thor-oughly wetted by the spray balls, but it does notensure that the tank will be cleaned by theplant CIP system.

Fixed sprayballs can normally achieve suc-cessful coverage test results in 30-90 secondsbased on a 15-20 GPM (3-5 m3/hr) flow and a ∆Pof 25 psi (1.7 bar) per spray ball (the number ofspray balls is determined by total flow require-ments and discussed later). The test is done byspraying a dilute (0.2-0.3 g /L) Riboflavin solu-tion onto vessel internals, allowing the Ribofla-vin to dry, and then using the spray balls toremove the residue. After the desired timeinterval, the tank is inspected with a UV lightto determine that the Riboflavin has been re-moved.

Ideally, the test would be successfully doneat 15 GPM (3 m3/hr) per spray ball and 30seconds to allow for a safety factor and providefor the possibility of increasing the number ofholes in the spray balls if the original drillingsdid not achieve the required coverage.

Because the basis of cleaning is the coveragetest results, it’s common to use a safety factorof 2-3 on the test time and use the same time forall steps in the CIP cycle. Based on six cleaningsteps of 5 minutes each and an allowance forset-up, air blows, heating, and chemical addi-tions, the time to clean a circuit containing amajor flow path (tank) and a few minorflowpaths (dip pipe or transfer line) is about 90minutes.

A thorough analysis of diversity is requiredto analyze all utility systems including CIP. A

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 49: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

face and reduce the differential between the microscopicpeaks and valleys. After electropolishing, the surface rough-ness is typically reduced by 50% to 15 Ra µ-in, but smoothingof the peaks is more important than the actual Ra value.

In addition to finish, careful attention must be given todetails such as dip pipes, agitator couplings, baffle attach-ments, and nozzle connections to eliminate pockets and deadends, provide smooth, crevice-free joints, and make equip-ment self-draining.

Flow Rate/TurbulenceTanksSpray balls are quite effective for cleaning equipment such astanks when properly designed for the particular application.Low-pressure spray is generally adequate since cleaning isperformed by a deluge/cascade/soak (chemical) action andnot by mechanical (impingement) force. The function of thespray balls is to distribute the washes and rinses to the top ofthe tank, wet all surfaces by a combination of spray andfalling film, and allow the chemical action to take place. Itmay be necessary to add a removable spray ball at a lowelevation to clean agitators, spargers, or side mounted nozzleslocated in the lower portion of a tank, but generally, topmounted spray balls are sufficient. The top spray balls shouldbe located a minimum of 6" above the highest liquid level toavoid the possibility of process fluids entering the spray ballwhen not in use and plugging the holes. Although somemanufacturers say they simulate a tank head and drill sprayballs with holes directed at specific nozzles, off-the-shelfspray balls generally provide adequate coverage with ap-proximately 1 spray ball for every 10-15 ft2 (1-1.5 m2) of crosssectional area.

Spray balls are typically sized for 15-20 GPM (3-5 m3/hr)each and a 25 psi (1.7 bar) ∆P. Performance is generally basedon a coverage test. The plan for coverage testing should bebased on the lower flow rate to allow flexibility. If fullcoverage is not obtained during the test, additional holes canbe drilled without affecting the allowed pressure drop. In-creased pressure is not desirable because it may cause atomi-zation, which would be detrimental as small liquid particleswould need additional time to coalesce and produce a clean-ing film on the tank walls.

It is best to operate multiple spray balls at the same time.Because of the flow-pressure characteristics of the centrifu-gal supply pumps, it is important to design all paths withina circuit for the same flow rate. This may require splitting theflow to spray balls. If this is the case, cycling betweenmultiple paths should be performed at frequent intervals, say30 seconds, to provide a reasonably consistent coverage ofinternals.

An estimate of the CIP flow rate for cleaning tanks isobtained by requiring a Reynolds Number > 2100 for the filmof fluid running down the tank walls. In Principles of Chemi-cal Engineering, studies show that with a given film viscosity,mass flow rate and wetted perimeter, Ref is the same whetheror not a cylinder is full.1

process simulation is performed to determine simultaneoususages of utility systems to ensure that adequate watersupplies are provided. The simulation is also used to deter-mine the number of CIP units required to operate simulta-neously which will affect both the diversity of operation andthe instantaneous need for Deionized Water (DIW) andWater For Injection (WFI).

TemperatureIf proteins are present, the pre-rinse should be done atambient temperature to remove as much protein as possiblewithout denaturation. Subsequent rinses and washes shouldbe done at higher temperatures, typically 140-180oF to im-prove solubility of other types of contaminants. The tempera-ture is typically raised with a sanitary steam-heated shelland tube heat exchanger.

Solution Concentration and TypeExcept for the final WFI rinse, the rinses and washes willconsist of ambient DIW. After the pre-rinse, the other solu-tions will be heated to 140-180°F. A detergent should beselected based on its ability to solubilize residue and the easewith which it can, in turn, be removed. The alkaline wash isusually made up to a 1-2% caustic concentration while theacid wash may have a slightly lower concentration of acid,typically phosphoric, to neutralize residual caustic and re-move calcium and magnesium carbonate deposits.

Satisfactory cleaning results can be obtained using a fairlywide range of chemical concentrations. However, to usecleaning solutions of different concentrations, it will be nec-essary to validate their efficacy over the range of concentra-tion expected.

Solutions can be made from commercially available (typi-cally food grade) bases and acids or proprietary solutions canbe purchased from firms specialized in cleaningbiopharmaceutical equipment.

Surface Characteristics of EquipmentHistorically, there has been disagreement as to the advantageof polishing compared to mill finish. The advocates of usingmill finish maintained that microscopic scratches providesurfaces for protein and other contaminant adherence. Al-though this may have been true for mechanical polishing, thisis not the case for electropolishing, where the sub-microscopic“scratches” are too narrow for contaminants to hide.

Internal FinishCleaning is a chemical rather than mechanical action. Sinceit’s important to minimize surface degradation caused bymechanical forces and/or chemical action, sufficient, but notexcessive chemical concentrations, temperature, and forceare applied to the surfaces being cleaned.

The typical biopharmaceutical finish is approximately 15Ra (Roughness Average) µ-in (0.38 µm) electropolished 316LSS. This is produced by mechanically polishing the mill finishto 25 Ra µ-in (0.6 µm) (max) surface roughness and thenelectropolishing. Electropolishing will both smooth the sur-

Page 50: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

mρ m

V = ______ = ______πDt ρπDt

πDtDH = 4RH = 4 × ______ = 4t

πD

DHVρ 4tmρRe f = ________ = __________________

ν ρπDt×0.000672µ

4mRe f = __________________

πD×0.000672µ

mRe f = 5952 ______

W µ

where: D Tank Diameter, ftDH Film Hydraulic Diameter, ftm lb mass per secondRef Film Reynolds NumberRH Film Hydraulic Radius, ftT film thickness, ftV velocity, ft/hrW vessel circumference, ftµ viscosity, cPv viscosity, lb/ft-secρ density, lb/ft3

This equation can be rearranged and rounded to produce:

GPM / ft = 2.5µ or m3/hr/m = 2µ

This relationship indicates that considerably less hot fluid isrequired, when compared to cold fluid, to achieve the samecoverage. For example, water viscosity is 0.35 cP at 180°F(80°C) and 1.0 cP at ambient conditions. In terms of absolutevalues, a 7'-6" diameter vessel would require 25 GPM (5.5 m3/hr) for a hot rinse and 75 GPM (17 m3/hr) for a cold rinse.

LinesDiffusion and convection are the controlling elements ofcleaning kinetics and indicate that flow need not be turbulentto clean the straight portion of a pipe. However, turbulencewill increase fluid movement to the surface where the solventcan mix and react with protein or other contaminants andalso assist in moving the resultant mixture away from thesurface.

For tube diameters > 1", a velocity of 0.5 ft/sec (0.2 m/s) issufficient to achieve turbulent flow. However, other consider-ations may govern the selection of a suitable velocity. Forexample, the velocity must be greater than the saltationvelocity to remove larger and heavier particles and highenough to entrain gas bubbles in case a portion of the line isnot self-venting. The higher velocity in the main run of thepipe also provides better flow into dead ended branches forcleaning when maximum distance criteria are followed. There-fore, the traditional recommendation (not a requirement) ofthe 3A–Dairy2 standard for a velocity of 5 ft/s (1.5 m/s) seems

Figure 1. Typical CIP system.

Page 51: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Tank No. of CIP TankDiameter Spray Balls GPM Line Sizes Outlet Size

3’- 4’ 2 40 11/2” 2”

4’-6’ 3 60 2” 21/2”

6’ - 10’ 4 80 21/2” 3”

Table A. CIP system design.

valid. This velocity should be based on the largest diameterwhen flowing through different diameter pipes or tubes inseries, but if there is more than a one line size change, thecircuit should be split to accommodate the different flowratesneeded to achieve similar velocities.

Design for CleaningPipingThreaded and flanged connections should not be used ascontaminants can accumulate in the threads or the pacebetween flanges and gaskets. Ideally, the system should becompletely welded.

Return lines should have as much slope as possible (pref-erably 2% but a minimum of 1%) to both encourage gravitydraining and discourage air pockets from forming whichwould prevent cleaning fluids from reaching the surfaces tobe cleaned. Pockets must be eliminated in design and fabri-cation by firmly supporting the lines to maintain the desiredslopes.

Branches should join the return header with minimumdead legs.3 Dead legs and pockets can retain dirt or evencleaning chemicals which will prolong the cleaning cycle orrequire additional rinse or wash fluids which increase boththe plant effluent and the DIW and WFI usage.

Lines should be cleaned individually or in series. Do notattempt to clean lines in parallel since it is difficult to ensurethat minimum velocities are achieved in each path and itmight be possible for one path to backup into another andactually impede cleaning.

No permanent connections should be provided betweenCIP and the process: transfer panels, hoses, or mix-proofvalves should be used to make and break connections. Usesanitary clamped connections at transfer panels and hoses:bolted for rarely opened and thumb screw for often opened.

Proximity switches or sanitary pressure gauges are usedto confirm paths and prevent mis-operation and/or unsafeoperation of the CIP circuit.

CIP SkidThe CIP skid normally contains a tank for rinse and chemicalsolutions. It also may contain a tank dedicated to WFI for thefinal rinse or WFI may be provided by a common tank tosupport several CIP skids. A circulating pump, heat ex-

changer, and chemical day tanks, along with piping andcontrols, complete the skid. Tanks should contain a means todisengage any air returned to the skid. Provide cleaning(spray) for CIP tanks since the tanks can be the dirtiest partof the system.

Return pumps can be mounted on the CIP skid if the skidis close to the process user(s), but this function is normallyprovided by pumps located close to the process equipment.The pump pressure and flow rate should be monitored toconfirm circulation rate.

FiltersSince filters are hard to drain, it may be necessary to remove,and possibly discard, the cartridge for cleaning and thenreinstall a new one before sterilization.

AccumulationEquipment and lines should be free draining to avoid a“bathtub ring” effect. Even over-sized bottom nozzles andvalves require a driving force of differential pressure toovercome dynamic losses. Return lines are typically under-sized to satisfy the minimum velocity requirements. Theresult will be accumulation in the tank, and there is apossibility that dirt will accumulate at the air-liquid inter-face. Outlet nozzles and lines should be sized to minimizeaccumulation using valve data and appropriate engineeringequations.

Accumulation or backup in a tank can be estimated byhydraulic calculations.4

GPMh = (___________)

2

19.636kd2

Valve Characteristic

GPM∆P = (________)

2

Cv

Combining these two equations and substituting k=0.61, thefollowing backup is required to flow through a given valve andnozzle.

GPM GPMh = (___________)

2

+ 28(___________)2

3.5d2 Cv

whereh = backup, ind = nozzle diameter, in

If there is accumulation in the target tank, the supply tankmay empty and stop the cleaning cycle until the accumulated

“Historically, there has been disagreement as to the advantageof polishing compared to mill finish.”

Page 52: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

volume drains back to the supply tank. When this happens,the cycle time will be extended, adversely affecting thereproducibility of the cleaning cycle. Rather than increase thequantity of cleaning chemicals, which may require additionalrinse volume and increase effluent quantity, the preferredapproach is to remove the restriction at the tank exit.

The combination of sprayball and line size criteria resultsare shown in Table A.

The process tank can be pressurized to provide the headrequired to overcome resistance in the outlet nozzle andvalve, but it is difficult to control the pressure to maintain theliquid level just at the outlet nozzle entrance. If the pressureis too low, accumulation will still occur, and if too high, therewill be additional air entrainment, which will reduce thecapacity of the return line and may interfere with cleaning.

There are some who suggest that starting and stopping theCIP feed pump can be used to remove accumulation, but thismerely makes the accumulation move up and down the tankwall. It does not eliminate the problem. It also requires asophisticated control system to keep track of the cleaningtime if the system is continuously stopped and started. Thesolution is to use a return pump to overcome the resistance ofthe tank outlet.

CIP SystemsIt is preferable to use a common philosophy for all CIPsystems in a plant. This will avoid operational errors, provideconsistent control system configuration, and maintain docu-mentation format between CIP systems and users.

A once-through system is the least costly system from acapital cost viewpoint, but the most costly to operate becausechemical solutions are made up, heated, then thrown away.

Systems use either a self-priming pump or an eductor toassist in returning and recycling washes and possibly somerinses to minimize chemical consumption, utilities, and efflu-ent. Drainage also will be improved with return pumpslocated close to the process equipment. The final rinse may ormay not be recovered to provide the pre-rinse for the nextcycle. Figure 2 shows a typical CIP skid system which in-cludes wash and rinse tanks, supply and return pumps, aheater, and all necessary, piping, instrumentation, and con-trols.

Eductor systems use eductors either alone or in combina-tion with pumps to assist with recirculation and to make upthe chemical solutions. They can pull vacuum and removetrapped air pockets and may be preferable to pumped returnssince they can’t lose prime. They also can provide moremotive force than a pumped system as they do not have tocontend with Net Positive Suction Head (NPSH) require-ments. Still an eductor can vapor bind with flashing fluids orentrained air.

Pump systems using low-speed, self-priming pumps havehigher flow rates and smaller diameter piping than eductorsystems. They have an advantage over eductor systems inthat they can be located at the target tanks to eliminateaccumulation. Both pump and eductor systems will operatebetter at colder fluid temperatures because of the lower vapor

pressure.Multiple circuits fed from the same CIP system should be

designed for the same operational flowrate. If both large andsmall equipment is cleaned using the same system withdifferent flow rates, there is the likelihood that both mini-mum and maximum velocity criteria will be violated in thepiping systems. Systems can be balanced by splitting sprayballs so that they are utilized either individually, in pairs, oreven two of three open at any one time to make all pathswithin a circuit use the same flow rate.

CIP CyclesDuring each stage of a CIP cycle, each moving componentshould be operated in the same sequence as during normaloperation. This includes valves, agitators, and pumps toensure each fluid successively contacts all surfaces. Typi-cally, each moving component would operate 5-6 times for 3-5 seconds during each step of the CIP cycle.

In order to minimize effluent and reduce chemical utiliza-tion, it may be possible to recirculate some portion of therinses. Perhaps the rinse can be drained for 1/3 - 1/2 of theallocated time and then recirculated for the remaining time.In addition, the remaining rinse can be used as the startingpoint for caustic and acid makeups.

CIP systems both remove contaminants and prepare equip-ment for steaming. A discussion of the nature of “dirt” and thechemicals used to remove it is beyond the scope of this article,but a brief synopsis of the solutions for a typical mammaliancell culture process is shown in Table B.

Typical steps in a CIP cycle are:

Pre-RinseThe pre-rinse uses either a fresh, clean, cool water (typi-cally DIW) source, or reuses the previous final rinse. The pre-rinse is used to remove residual process fluid and debris. Thefluid and temperature should be selected to avoid denatur-ation and precipitation of proteins. As noted earlier, this stepmight be once through followed by recirculation.

Recirculated Alkaline WashResidual rinse water could be heated and fed with caustic or

Figure 2. CIP skid.

Page 53: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 3. 21 CFR Parts 210 and 211.

(a) Equipment and utensils shall be cleaned, maintained, and sanitized atappropriate intervals to prevent malfunctions or contamination that wouldalter the safety, identity, strength, quality or purity of the drug productbeyond the official or other established requirements.

(b) Written procedures shall be established and followed for the cleaning andmaintenance of equipment, including utensils, used in the manufacture,processing, packing, or holding of a drug product. These procedures shallinclude, but are not necessarily limited to, the following:

(1) Assignment of responsibility for cleaning and maintaining equipment;

(2) Maintenance and cleaning schedules, including, where appropriate,sanitizing schedules;

(3) A description in sufficient detail of the methods, equipment, andmaterials used in cleaning and maintenance operations, and the methodsof disassembling and reassembling equipment as necessary to assureproper cleaning and maintenance;

(4) Removal or obliteration of previous batch identification;

(5) Protection of clean equipment from contamination prior to use;

(6) Inspection of equipment for cleanliness immediately before use;

(c) Records shall be kept of maintenance, cleaning, sanitizing, and inspectionas specified in §§211.180 and 211.182.

Solution Purpose

Water - 1st rinse gross removal of contaminants

Base with Hypochlorite Solubilize and denature proteins

Water - 2nd rinse Remove base and debris

Acid Neutralize baseDissolve mineral saltsPassivate

Water - 3rd rinse Remove acid and debris

WFI Rinse Remove all remaining contaminants

Steam Destroy pathogens

Table B. Chemical solutions form CIP/SIP.

other detergent to make up the alkaline (typically 1-3%caustic) wash. The step uses the alkaline wash to denatureand solubilize the remaining proteins. Although denatur-ation increases the dirt load and makes proteins more diffi-cult to remove, the bulk of the easy-to-remove proteins shouldhave been removed in the pre-rinse step. If desired cleaningresults are not obtained, this is the step which normallyprovides the most benefit from an increase in time.

Proprietary chemical cleaners are available for both alkaliand acid washes. These mixes may contain a mixture ofchemicals, detergents, chlorine, or other additives to improvethe cleaning action.

Air blows are used after chemical washes to maximizechemical removal and make the succeeding rinse easier.

Hot RinseThis rinse is used to remove the alkalinity and additionaldirt. This step will not remove (solubilize) additional proteinand may be recirculated or once through.

Recirculate Acidified WashIf necessary, an acid wash is used to neutralize residual base,solubilize remaining dirt (inorganic), remove mineral depos-its, and passivate the surface. It is sometimes omitted and itmay be made up from the residual rinse fluid from theprevious step.

Hot RinseResidual acid and any additional dirt loosened in the acidwash is removed with a hot DIW rinse. This rinse also may berecirculated.

Final RinseA final rinse with WFI removes traces of previous wash. It ismonitored with pH, conductivity, or resistivity (compared toinlet) to ensure cleaning by measuring removal of chemicalsolutions. These variables will not detect protein residues.

ControlSome companies believe that it is easier to validate a manualCIP procedure because the control system is not involved inthe validation procedure. However, current control systems,when properly implemented, have many advantages over a

manual system and perhaps the ideal situation is a combina-tion of manual and automatic control. It is important that thecontrol system be easy to monitor, control, and validate.

Automated CIP is the most consistent method to achievereproducible cleaning. The use of a control system will ensurethat cycles, duration, and sequence objectives are achievedeach time a CIP is performed.

The main control elements are time, temperature, andflow rate. Cleaning is usually verified by monitoring rinseconductivity. Measurements are made and recorded to verifythat achievable tolerances consistent with the cleaning objec-tives are achieved. Typical operating tolerances of chemicalsolutions concentration is 1-3%, temperature accuracy shouldbe within ±5oF (3°C), and a flow rate variation should notexceed ±10%.

The main function of the control system is to pulse valvesand operate rotating equipment within a circuit with eachrotating or moving component cycled 5-6 times during eachstep of the cleaning cycle. Communication with the processcontrol system is essential to coordinate the cycling of path-ways in the process circuit with the change in CIP steps toverify that the various operations occur for the desired timeat the proper temperature and composition. Other controlfunctions include chemical addition rate and concentrationand temperature control.

Once a control system is employed, it is necessary toconsider what action(s) to take in the event of a deviation. Onepotential deviation is an external requirement to stop thesystem because of a failure such as a hose leak. Anothercommon problem occurs when low level occurs in one of thetanks on the CIP skid and the circuit must be put in recycleto avoid damaging the pump. The CIP system status can beconsidered as one of three or four states.

Page 54: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

CIP System Design

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

1. Normal - target circuit cycles valves to clean associatedpaths.

2. Makeup - main path in targeted circuit is open for heatupand chemical addition.

3. Hold - drain valves are closed to prevent loss of chemicals,valves are not cycled, and timer is paused in the CIP PLCuntil state is returned to normal. This state may be thesame as makeup.

4. Abort - systems fail to pre-established safe position.

A control system should be used to validate cleaning. Inaddition to the controls already discussed, the system shouldprovide for documenting the results of each cleaning opera-tion.

ValidationCIP systems provide for reproducible uniform cleaning andminimize the possibility of human error. The cost of mainte-nance is reduced, down time is minimized, and operatorsafety is improved.

With proper documentation, the system can be easily andquickly validated to prove that the cleaning was effective andcleaning agents have not been introduced into the product.

In order to validate a CIP system, written documentationis required to define the procedures for cleaning each piece ofequipment, circuit, and flow path. Additional procedures arerequired to quantify the cleanliness required by the process,a means to measure the cleanliness or residue, and ananalytical method to confirm the measurement results. Theprotocol which describes the validation procedure for CIPmust be reviewed, approved, and executed by technicallycompetent personnel.

Figure 3 is an excerpt from 21 CFR-Parts 210 and 211.3

ConclusionProper implementation of CIP seems to be a mixture of artand science. There is nothing wrong with using empiricalrelationships provided they don’t conflict with good engineer-ing practices. A well-designed CIP system can achieve mini-mum flow velocities in both supply and return lines withoutaccumulating fluid in the tanks being cleaned. A combinationof properly designed outlet connections and return pumpscan be used to achieve the desired results.

References1. Principles of Chemical Engineering, Walker, Lewis,

McAdams, and Gilliland, McGraw Hill, 1937, 3rd Ed, p.113.

2. International Association of Milk, Food, and Environmen-tal Sanitarians.

3. ASME BPE-2002, The American Society of MechanicalEngineers, 2002.

4. Cameron Hydraulic Data®, Ingersoll-Rand Co., 1970, p.67.

About the AuthorDavid Greene is Process Director for theAker Kvaerner Biopharmaceutical BusinessUnit. He has been with Kvaerner for 25 yearsand has been responsible for the processdesign of biotechnology facilities for the past20 years. He has designed a wide variety ofindustrial microbial and mammalian cellculture facilities for clients in the United

States and Europe. Greene has a BSChE from the Universityof Rhode Island and an MBA from Northeastern University.He is a registered Professional Engineer in MA and NJ anda member of AIChE.

Aker Kvaerner, 440 Route 22 E., Bridgewater, NJ 08807.

Page 55: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Passivation

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

The Effect of Passivation on AL-6XNAlloy Compared to the Traditional304L and 316L Stainless Steel Alloys

by Arnie Grant and John Jermain

This articledescribes theresults ofsurfacecompositionalanalysis usingAuger ElectronSpectroscopyandelectrochemicalcorrosion testingof passivatedversusunpassivatedAL-6XN, 304Land 316L. First patented in 1985 by Allegheny

Ludlum, AL-6XN was designed to be ahigh strength, corrosion resistant “su-per-austenitic” alloy intended for ser-

vice in seawater. The alloy’s exceptional corro-sion resistance has led to its use in a largevariety of industrial applications, and mostrecently in the pharmaceutical/biotech indus-try. Given the inherent high degree of corrosionresistance, the question has been raised as tothe need for passivation of this alloy. Thisarticle describes the results of surface composi-tional analysis using Auger Electron Spectros-copy and electrochemical corrosion testing ofpassivated versus unpassivated AL-6XN, 304Land 316L. A proprietary chelant passivationformulation and process was employed. Cr/Feratio in the passive layer and pitting potential(Epit) were the criteria used to assess the effec-tiveness of the passivation.

Introduction“Pharmaceutical equipment and high puritywater systems are designed so that product

contact surfaces are not reactive, additive, orabsorptive so the drug product is not adverselyaltered.1” Austenitic stainless steel alloys, pri-marily 316L and much less frequently 304L,either mechanically polished or electropolished,have been the metal alloy of choice for piping,tubing, fittings, tanks, and equipment becauseof these requirements. Factors such as cost,physical and mechanical properties,fabricability/weldability, compatibility withprocess, product cleaning and sterilizing fluids,and corrosion resistance must be considered.

Unfortunately, even 316L suffers from fre-quent and extensive corrosion in the forms ofrouging or pitting in aggressive environmentsincluding clean steam, hot purified water/HWFI, and the various product and intermedi-ate fluids. Rouging2-4 may be the result of gen-eral corrosion, where the metal corrodes at aconsistent rate over the entire surface, or therouge may originate elsewhere and migrateand deposit at the observed location. Pitting isa highly localized attack, in which only a smallarea of the metal surface is affected, but the

Figure 1.PAMO meterand test cell.

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 56: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Passivation

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

rate of corrosion in this small area is moderately high. Inthose circumstances where the degree of corrosion that isoccurring is unacceptable, AL-6XN has become one of the newalloys of choice. Lifecycle cost comparisons are beyond thescope of this article.

The chemical compositions and relative approximatecosts (with 304L taken as 1) for 304/304L, 316/316L, andAL-6XN are listed in Table A.5-7 The most common grade andleast expensive is Type 304/304L, which makes up morethan 60% of all the stainless steel made in the United Statestoday. The improved corrosion resistance of the other gradesof stainless steel in this study is developed by addingexpensive alloying elements such as chromium, nickel, andmolybdenum.

Alloy: Type 304LThe most common grade of austenitic stainless steel producedin the United States is type 304/304L. Type 304L is upgradedfrom type 304 by slightly increasing the percent composition ofnickel and lowering the carbon composition. In this low-carbonaustenitic alloy, control of carbon to a maximum of 0.03% hasbeen shown to minimize carbide precipitation (sensitization)during welding and concomitant susceptibility to intergranu-lar corrosion in the Heat Affected Zone (HAZ).

Alloy: Type 316LBy adding 2 - 3% molybdenum, type 316 has significantlybetter chloride corrosion resistance. Type 316 is upgraded bylowering the allowable carbon content to reduce/eliminatesensitization and slightly increasing the nickel composition.By containing molybdenum and increasing the percentagesof nickel, this alloy displays higher strengths at elevatedtemperatures and better corrosion resistance than the 304austenitic alloys.

Alloy: AL-6XNAL-6XN was initially intended to be used in a seawaterenvironment, but extensive testing has demonstrated it to beresistant to a variety of corrosive elements. Its excellentchloride pitting resistance is attributable to its 6.50% molyb-denum content, while its significant resistance to chloridestress corrosion cracking is a result of its nickel content ofabout 25.00%. The addition of nitrogen enhances its pittingresistance as well as mechanical strength. Nitrogen alsoserves to significantly reduce the formation of potentiallyharmful secondary phases during the manufacture of largecross-section products. The AL-6XN alloy was tested againstother stainless steel alloys, and it was concluded that it is themost corrosion resistant iron-base austenitic stainless alloy

Table A. Composition of stainless steel alloys.

Composition Stainless Steel Alloys

304 304L 316 316L AL-6XN

Carbon 0.08 % 0.035 % 0.08 % 0.035% 0.03%

Chromium 18.00 / 20.00 % 18.00 / 20.00 % 16.00 / 18.00 % 16.00 / 18.00 % 20.00 / 22.00 %

Copper ----- ----- ----- ----- 0.75 %

Iron 66.00 / 71.00% 64.00 / 70.00 % 62.00 / 69.00 % 61.00 / 68.00 % 42.00 / 47.00 %

Manganese 2.00 % 2.00 % 2.00 % 2.00 % 2.00 %

Molybdenum ----- ----- 2.00 / 3.00 % 2.00 / 3.00 % 6.00 / 7.00 %

Nickel 8.00 / 11.00 % 8.00 / 13.00 % 10.00 / 14.00 % 10.00 / 15.00 % 23.50 / 25.50 %

Nitrogen ----- ----- ----- ----- 0.18 / 0.25 %

Phosphorus 0.045 % 0.040 % 0.045 % 0.040 % 0.040 %

Silicon 1.00 % 0.75 % 1.00 % 0.75 % 1.00 %

Sulfur 0.030 % 0.030 % 0.030 % 0.030 % 0.030 %

Relative Cost ----- 1.0 ----- 1.2 – 2.5 3.5 – 5.0

Table B. Passivation monitor results for alloy tube samples.

Stainless Alloys Method of Testing Non-Passivated Samples Passivated Samples

AL-6XN Passivation Monitor 993 mV 1008 mV987 mV 1002 mV

Average 990 mV 1005 mV

304L Passivation Monitor 27 mV 57 mV26 mV 52 mV

Average 27 mV 55 mV

316L Passivation Monitor 55 mV 106 mV56 mV 113 mV

Average 56 mV 110 mV

Page 57: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Passivation

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

presently available. According to CSI6, unless a system isconstructed entirely of AL-6XN, special precautions must betaken to avoid potential galvanic corrosion problems.

Evaluation of PassivationThe passivation acceptance or inspection methodology forstainless steel typically found in government or associationspecifications such as ASTM A-380, MIL-STD-753, QQ-P-35,etc., are sensitive colorimetric spot tests to confirm thecomplete removal of corrosion inducing surface contamina-tion by free iron or carbon steel and timed exposure tests inwater, high humidity, or salt spray for the onset of visiblecorrosion. The acceptance criterion for the time to onset ofcorrosion is left to the discretion of the facility owner or hisdesignated authority. No other criteria for evaluating passi-vation are noted.

Recently, more sophisticated instrumental techniques havebeen employed to characterize the chemistry of the very thin(30 to 50 Angstrom) passive layer by accurately measuring theremoval of iron and the enrichment of chromium, which hasbeen shown to have a direct correlation to improved corrosionresistance. In the past, an improvement in the Cr/Fe ratio fromthe nominal bulk composition ratio of approximately 0.25 to avalue of Cr/Fe = 1.5 has been the unofficial benchmark targetfor passivation of 316L. No similar criteria have been found for304L or AL-6XN. X-Ray Photoelectron Spectroscopy (XPS/ESCA) and Auger Electron Spectroscopy (AES/SAM) are thetwo analytical techniques employed to measure the elementalcomposition as a function of depth from the surface, throughthe passive layer and into the bulk alloy.

In addition, electrochemical corrosion measurement tech-niques have been used to evaluate the effectiveness of passi-vation by accurately measuring the corrosion resistance, e.g.,pitting potential (ASTM G-61) or Critical Pitting Tempera-ture (ASTM G-150). As in other passivation studies by theauthors8,9, both AES to measure chromium enrichment/ironremoval in a Cr/Fe Ratio Depth Profile and CyclicPotentiodynamic Polarization to measure pitting potential(Epit) have been employed to evaluate passivation

MethodologyMaterialThe stainless steel tubing used in this study was donated byCentral States Industrial. The AL-6XN tubing was identifiedas 2.0" O.D. X.072, 15 GA, ASTM A270, Polished, Heat

#882648. The 316L was identified as 2.0" O.D. X.065, 16 GA,ASTM A270, Polished, Heat #19126. The 304L was 2.0" O.D.X.065,16 GA, ASTM A270, Polished. The Heat # was notspecified. The tubing was cut into 2.0" lengths for the electro-chemical test coupons, while the AES test coupons were ¾"wide longitudinal segments.

Passivation ProcedureAfter the test coupons were cut, square faced, and deburred,all samples were first cleaned in an alkaline detergent/chelant treatment and then passivated per a standard, pro-prietary procedure.

Electrochemical Passivation Monitor (Pamo Meter)The pitting potentials of the three stainless steel alloys,304L, 316L, and AL-6XN, were measured in duplicate usingthe electrochemical Passivation Monitor (PAMO) shown inFigure 1 with the test cell configuration. The electrolytesolution was 0.5 M KCl, the test temperature was 25°C, thecounting electrode was 316L, and reference electrode wasCalomel. The field portable monitor was developed in con-junction with the Materials Science Department at USC toprovide more rapid data on pitting potential that was essen-tially equivalent to the more time consuming ASTM G-61.The procedure measures the potential in millivolts at whichelectrochemically induced pitting occurs. The pitting poten-

Figure 2. Pitting potentials of 304L, 316L & AL-6XN stainlesssteel alloys.

Table C. Measurements from auger electronspectrometer.

Sample Oxide Thickness Maximum Cr/Fe Cr/Fe Cr EnrichedIdentification (Å) Ratio Ratio @ 10Å Layer

Unpassivated AL-6XN 150 1.50 1.52 38.7

Passivated AL-6XN 100 3.03 2.56 82.2

Unpassivated 304L 44 0.38 0.24 NONE

Passivated 304L 33 1.46 1.25 13.3

Unpassivated 316L 80 0.25 0.08 NONE

Passivated 316L 24 1.66 1.12 11.3

Page 58: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Passivation

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 3. Cr/Fe ratio depth profiles for Type 304L.

Figure 4. Cr/Fe ratio depth profiles for Type 316L. Figure 5. Cr/Fe ratio depth profiles for AL-6XN.

tial is measured after fifteen minutes of an applied current tothe test cylinder via the potentiometer as compared to ap-proximately six hours for the G-61 procedure. The pittingpotential, Epit, value may then be conveniently used to com-pare corrosion susceptibility before and after passivation forthe same alloy as well as comparison among the differentalloys.

Auger Electron Spectroscopy (AES)AES element concentration and Cr/Fe Ratio Depth Profileswere performed employing a Physical Electronics Spectrom-eter with interfacing and software upgraded. Operationalparameters for the quantitative elemental analysis were asfollows:

Parameters:Beam Energy: 5000 VMagnification: 2500XBeam Current: 1.4 µAIon Etch Rate: 60Σ/MIN Ta2O5

Experimental ResultsResults from Electrochemical Passivation MonitorThe pitting potential data for the three alloys are presentedin Table B and shown graphically in Figure 2. When tested

against the traditional 304L and 316L stainless steel alloys,AL-6XN’s pitting potential is significantly higher in bothpassivated and non-passivated conditions. When comparingthe pitting potentials of the traditional stainless steel alloysbefore and after passivation, the pitting potentials doubledfor both the 304L and 316L. In contrast, the AL-6XN alloydisplayed only a very slight increase in pitting potential afterpassivation.

Results from Auger Electron SpectroscopyThe AES data are tabulated in Table C, while the Auger Cr/Fe Ratio and element concentration Depth Profile curves arepresented in Figures 3 through 8. In all cases, the passivatedsamples had a much higher chromium to iron ratio then thesamples that were not passivated.

DiscussionWhen compared to 304L and 316L stainless steel alloys, theAL-6XN pitting potential is significantly higher (an order ofmagnitude and greater) in both non-passivated (990 mV) andpassivated (1005 mV) conditions, confirming once again itsoutstanding pitting corrosion resistance. When comparingpitting potentials of the traditional stainless steel alloys, thepitting potential doubled for both the 304L (from 27 mV to 55mV) and 316L (from 56 mV to 110 mV) after passivation.

Examination of the Auger elemental analysis data pro-vides a somewhat different picture. The two primary objec-tives of passivation are 1) to remove any surface contamina-tion which may promote corrosion, and 2) to remove iron toenhance the Cr/Fe Ratio values. These two goals are illus-trated in the Cr/Fe Ratio Depth profile curves. In theunpassivated condition, all three alloys display the lowest Cr/Fe ratios at the surface in the first 10 Angstroms, risingsharply to a peak at approximately 10 to 20 Angstroms. Thisis taken as an indication of surface contamination. As ageneral rule, it is desirable to have the maximum Cr/Fe valueas close to the surface as possible. Not only does Cr/Feincrease dramatically after passivation (304L from 0.38 to1.46; 316L from 0.25 to 1.66; and, AL-6XN from 1.50 to 3.03),but the peak values are at or very near the surface, indicatingboth objectives have been met. The depth profile curves also

Page 59: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Passivation

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

show that, at the surface of the three unpassivated alloys,iron is seen to be higher than chromium although onlyslightly for the AL-6XN. After passivation, a dramatic shift isseen where chromium is much greater in concentration thanthe iron. The chromium enriched layer value shown in TableC represents the cross-over point, the depth to which the Cris greater than Fe. For AL-6XN, the enrichment depth isdoubled by passivation, from 38.2 Angstroms to 82.2 Ang-stroms.

Should 304L and 316L alloys be passivated? Without adoubt. Should AL-6XN be passivated? The electrochemicalpitting test results do not show a clear need based on thedifference in Epit before and after passivation. However, giventhe significant removal of iron from the surface of the AL-6XNby the passivation process and the certain requirement for athorough cleaning to remove surface contaminants afterinstallation, it seems most prudent and cost effective torequire both cleaning and passivation for new AL-6XN sys-tems. Evaluation of the effect of welding on surface segrega-tion of alloy constituents in AL-6XN and the need for passi-vation is currently underway. As experience is gained in the

operation of equipment constructed of AL-6XN in pharma-ceutical applications, we will be able to determine if rougingor corrosion occurs, under what conditions and frequency,and if maintenance derouging and repassivation will berequired.

References1. ISPE Water and Steam Systems Baseline® Guide, Janu-

ary 2001.

2. Self, T., Oslen, P., and Banes, P., “Investigating theRouging of Stainless Steel USP Water Systems,”Microcontamination, May, 44-55 (1993).

3. Menon, G.R., “Rouge and Its Removal from High-PurityWater Systems,” BioPharm, 3 (6), 40-43 (1990).

4. Tverberg, J. and Ledden, J., “Rouging of Stainless Steel inWFI and High Purity Water Systems,” Personal Commu-nication, 1998.

Figure 6a. 304L Unpassivated. Figure 6b. 304L Passivated.

Figure 7a. 316L Unpassivated. Figure 7b. 316L Passivated.

Page 60: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Passivation

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

5. Suzuki, O., Newberg, D., and Inoue, M., “Discolorationand its Prevention by Surface Treatment in High-PurityWater Systems,” Pharmaceutical Technology, April 1998.

6. Stainless Steel Selection Guide - Central States Industrial,Central States Industrial Equipment and Services Inc.,Springfield, MO., 2001.

7. Grubb, J.F., AL-6XN ALLOY - Allegheny Ludlum Corpora-tion, Allegheny Teledyne Company, Pittsburgh, PA., 1995.

8. Annual Book of ASTM Standards - A 270, AmericanSociety for Testing and Materials, Philadelphia, PA.,1990.

9. Grant, A., Henon, B., PhD, and Mansfeld, F., PhD, “Ef-fects of Purge Gas Purity and Chelant Passivation on theCorrosion Resistance of Orbitally Welded 316L StainlessSteel Tubing,” Pharmaceutical Engineering, March/April 1997.

10. Grant, A., “Passivation of Welds on Stainless Steel Tub-ing,” Presented at the ASME Bioprocess EngineeringSymposium, San Francisco, California, October, 1995.

AcknowledgmentsAlloy tubing was provided by Ken Kimbrel at Central StatesIndustrial Equipment and Services Incorporated, Spring-field, MO. Auger Electron Spectroscopic analyses were per-formed by Dr. David Dahlgren at PhotoMetrics Incorporated,Huntington Beach, CA.

About the AuthorsArnie Grant is Director of Research at Cal-Chem Corp., an international service organi-zation providing precision cleaning and pas-sivation to the pharmaceutical industry. Hereceived a BS in chemistry from FairleighDickinson University and pursued post-graduate courses at UCLA and CaliforniaState University. He has more than 35 years

of experience in analytical chemistry methods development,corrosion prevention and control, parts materials and processevaluation, and contamination control and prevention. Hewas past chairman of the ICRPG Infrared Committee and iscurrently active in ISPE and ASME presenting papers andconducting seminars in passivation and corrosion.

John Jermain is a research technician atCal-Chem Corp., an international serviceorganization specializing in precision chemi-cal cleaning and passivation for thebiopharmaceutical industry. Jermain is cur-rently enrolled in the chemistry program atthe California Polytechnic University atPomona and plans on pursuing post-gradu-

ate courses in business and law.Cal-Chem Corp., 2102 Merced Ave., So. El Monte, CA

91733.

Figure 8a. AL-6XN Unpassivated. Figure 8b. AL-6XN Passivated.

Page 61: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

Quality Systems in thePharmaceutical Industry

by Lorna Foote

This articlediscusses amethod adoptedby IT SoftwareSuppliers tomeet afundamentalrequirement ofall IT users - toreceive a goodquality productor service.

Within the pharmaceutical world,quality and validation require-ments for software products arefairly well covered and understood.

IT directors, their managers, and staff knowthat if they are implementing IT systems in aregulated area, they have to validate thesesystems and the systems and associated docu-mentation have to meet certain quality re-quirements. There are well-documented rea-sons for carrying out the validation and meet-ing the quality requirements, and there arewell-documented methods and standards de-scribing the goals and activities. There maystill be areas where these methods and stan-dards are not rigorously applied; for example,how many end-user developed databases be-come business critical, but have had little test-ing and have no documentation? However, nomatter how well known these issues are withinthe pharmaceutical industry now, the pharma-ceutical customers’ requirements will change,and the capabilities of the suppliers will changewith time.

The suppliers of IT products and servicesare constantly changing; they are gaining newskills, broader experience, answering the chal-lenges of the technology, and meeting the needsof competition. But, through all that change dothey continue to meet the quality and valida-tion requirements of the pharmaceutical in-dustry?

This article will discuss one method beingincreasingly adopted by IT software suppliersto meet a fundamental requirement of all ITusers – to receive a good quality product orservice. That method is the Software Engi-neering Institute Capability Maturity Model(SEI CMM). The Carnegie Mellon SoftwareEngineering Institute Web site describes CMMas follows:1

“The Capability Maturity Model for Soft-ware describes the principles and practicesunderlying software process maturity and isintended to help software organizations im-prove the maturity of their software processesin terms of an evolutionary path from ad hoc,chaotic processes to mature, disciplined soft-ware processes.”

It is reasonable to speculate that supplierswho have mature, disciplined, proven, and cer-tified software processes will deliver softwareof a high quality. To elaborate on that specula-tion, and to argue that this will help softwaresuppliers meet the regulatory needs of ourindustry, this article will briefly summarizethe CMM and its levels of maturity. Addition-ally, the article will draw comparisons betweenthe processes required to achieve the higherlevels of CMM and the processes required bydisciplines such as ISO 9001 and the ISPEGood Automated Manufacturing Practice(GAMP) Guide. These comparisons are neithereasy nor obvious since there is little directcorrelation between the elements of the CMM,the ISO standard, and the GAMP Guide. Infact, the purpose of each is so different that thebasis of comparison may at first seem question-able. However, it is hoped that the way in whichthe comparison is documented will aid an un-derstanding of how high levels of quality insoftware can be achieved through the CMMapproach. It should be noted here that there aremany standards, guides, and regulations thatcould have been used for this comparison; how-ever, the GAMP Guide is included because it isspecific to meeting the requirements of thehealthcare industry for automated systems andISO 9001 is arguably the best known standardagainst which quality systems are compared.2,3

So, the article aims to show how a very highproportion of the pharmaceutical industry’s

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 62: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

quality and validation requirements for the supply of soft-ware can be an integral part of the delivery process, evenwhen the specifics of the pharmaceutical industry’s regula-tory requirements are not explicitly addressed. The aim is notto suggest that we shift our focus toward the CMM, instead ofGAMP or ISO 9001, rather that it is an additional way oflooking at the quality of software suppliers, giving the phar-maceutical industry a broader choice of potential suppliers.

For those readers well versed in validation requirementsand processes, the following definition will be familiar; how-ever, it is included here as a reminder of the objective ofvalidation.

Validation: “Establishing documented evidence which pro-vides a high degree of assurance that a specific process willconsistently produce a product meeting its pre-determinedspecifications and quality attributes.”4

The Capability Maturity Model has been designed to helpsoftware developers achieve the consistency of quality that isrequired by validation. However, it must be stressed herethat the Capability Maturity Model does not in any waydirectly address the requirements of validation, since theyare understood within our industry. The introduction to oneof the main CMM texts states that the “CMM guides softwareorganizations that want to gain control of their processes fordeveloping and maintaining software and to evolve toward aculture of software engineering and management excellence.”It will help organizations identify the critical areas to addressto achieve the improvement in their processes and in thequality of their software products.5

This article will demonstrate that by implementing aCMM approach to software engineering processes or bycontracting suppliers who have achieved the higher levels ofCMM, systems will:

• be engineered according to defined and documented pro-cesses

• be delivered with documented evidence• provide a high degree of assurance• work consistently• meet pre-determined and agreed requirements and speci-

fications• meet pre-defined quality attributes• while this does not mean that the systems are validated

it will provide us with a system that can be validated.

The next two sections will briefly describe the background toand the objectives of the GAMP Guide and ISO 9001 respec-tively. The following section will describe the CMM in muchmore detail. This is intentional since this article sets out tointroduce the model which is considerably less well knownwithin the industry.

The GAMP GuideThe GAMP Guide was originally a set of guidelines forsuppliers of automated systems to the pharmaceutical indus-try. It was the work of the GAMP Forum, an industry groupset up to promote the understanding of validation of auto-

mated systems, and was first published in 1994. Furtherrevisions were published in 1996, 1998, and GAMP 4 waspublished in 2001. This most recent publication has beenrevised and refined to include current regulatory expecta-tions and good practice and now aims to provide guidance notjust to the pharmaceutical sector, but also to related healthcareindustries.

The GAMP Guide is an excellent source of informationabout what has to be done to validate a system. This includeswhat the end-user or owner of the system must do and whatthe supplier should do. The italicized and emboldened em-phasis in the last sentence is intended to show two things.The first is to place responsibility clearly with the owner/end-user who must make the decisions about how and if thesystem can be used for their purposes. The second is toindicate that should the supplier fail to meet some of therequirements of validation normally associated with a sup-plier, the owner/end-user would then have to, through extraeffort, provide the assurances that the system is satisfactoryfor use.

Perhaps it is because the responsibility for validation liesfairly and squarely in the hands of the system owner/end userthat the GAMP Guide is well known within the pharmaceuti-cal industry and much less known by “generic” IT suppliers.And, perhaps this situation will not and need not change.However, there are two downsides associated with IT suppli-ers not understanding the requirements of system validation.

• Some IT suppliers lose out because, although they may bemore than capable of supplying systems meeting thequality and validation requirements, they do not have thespecific knowledge of validation, its terminology, or theapplicable regulations. To make matters worse, they maynot be told about them as part of the tendering or procure-ment process, and so they miss out on opportunities tosupply.

• Some pharmaceutical companies lose out because, under-standably, they tend to look for suppliers who are knowl-edgeable about validation and the applicable industryregulations. This limits the source of suppliers by leavingout many IT companies supplying goods and services to aquality that could more than satisfy the industry’s needs.

It is not the intention to try to summarize the GAMP Guide– it is beyond the scope of this article. However, below is a listof points particularly pertinent to this article.

• In order to play their part in the production of a validatedsystem, it is recommended that the suppliers follow aformal management system, preferably based on stan-dards such as ISO 9000 Series.

• The supplier’s management system should follow a stan-dard life cycle approach which can be tailored as appropri-ate to the system being produced.

Page 63: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

• Contractually agreed Quality Plans and Project Plansshould be produced and meet the minimum standardsspecified in the Guide.

• Specifications should be produced for the required func-tionality, design, build, and finally, for the testing of thesystem. These also should meet minimum standards andthey should be formally agreed.

• The supplier should carry out testing and this should besufficient to show that the product is fit to be installed andused at the customer’s site.

• A further phase of testing carried out at the customer’s siteshould demonstrate the system meets the customer’srequirements.

While the Guide does include some detail concerning thesections and content that should be contained in a formalmanagement system, it clearly leaves the supplier with scopeto build a management system appropriate to their specificways of working and product range. Allowing this autonomyis essential; otherwise, the range of suppliers with which thepharmaceutical companies could work would be even moreseverely limited.

One of the real strengths of the Guide (GAMP 4 in particu-lar) is that it provides the pharmaceutical company or thesupplier to the pharmaceutical company a number of veryuseful Guidelines and Procedures that can be adopted andadapted to meet the specific processes of the organization.This approach (describing “how” as well as noting “what”) isone of the main differences between the GAMP Guide andISO 9001 and the CMM.

ISO 9001ISO 9001 is a “model for quality assurance in design, devel-opment, production, installation, and servicing.” It was for-merly British Standard BS 5750 and is now a European andinternational standard. BS EN ISO 9001 was published in1994 and there is now a new ISO 9001:2000. It is one of threestandards dealing with quality system requirements that canbe used for external quality assurance purposes. These stan-dards are generic and independent of any specific industry oreconomic sector.

BS EN ISO 9001: 1994, which was used in the preparationof this article, consists of 20 quality system requirements,each of which is expanded giving a total of 46 subsectionsdefining requirements. The objective of these requirementsis to help a supplier meet customers’ expectations and therebyachieve customer satisfaction.

Organizations seeking to demonstrate compliance withthe standard are likely to go through a registration processwhich will typically involve the following four steps:

• pre-audit to determine areas where practices are fallingshort of the audit - an optional stage likely to be done if theorganization is not confident of their ability to meet thestandard

• audit - done by an independent, qualified, and certifiedauditor. This will result in a pass or fail status.

• registration - in the event that the audit status is pass theorganization can progress registration

• surveillance audit - to determine the continuing state ofthe organization in their attempts to comply with thestandard

SEI CMMThe Software Engineering Institute started the developmentof a maturity framework in 1986 with assistance from theMITRE Corporation. It was initiated because the US federalgovernment needed a method of assessing the capability ofsoftware suppliers. This evolved by the early 1990s into theCapability Maturity Model, which is based on the actualpractices of organizations looked at during the developmentof the model. The model reflects the best of those practicesand is both fully documented and publicly available. TheCMM has continued to evolve with the introduction of addi-tional models (looking wider than the software engineeringprocesses) and a new integrated model. Because there isFigure 1. Structure of the Capability Maturity Model.

Page 64: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Related GAMP or ISO Requirement

GAMP – Planning – Validation Plan and User Requirements SpecificationGAMP – Design Review and Traceability MatrixGAMP – Functional Specification

ISO – Contract Review – to determine if requirements are adequatelydefined, agree with the bid, and can be implemented.

GAMP – Planning – Quality and Project PlansGAMP – Risk AssessmentGAMP – Categorization of Software and Hardware

ISO – Management ResponsibilityISO – Quality SystemISO – Contract ReviewISO – Design ControlISO – Process Control

GAMP – Quality and Project PlansGAMP – Project Change Control

ISO – Management Responsibility

GAMP – Subcontractor ControlGAMP – Supplier Audit

ISO – PurchasingISO – Control of Customer Supplied Product

GAMP – Quality & Project PlansGAMP – ReviewsGAMP – Good Engineering PracticeGAMP – Production, Control and Review of SoftwareGAMP – Design SpecificationsGAMP – Testing

ISO – Management ResponsibilityISO – Quality SystemISO – Process ControlISO – Corrective and Preventative actionISO – Internal Quality Audits

GAMP – Configuration ManagementGAMP – Document Management

ISO – Design ControlISO – Document and Data ControlISO – Product Identification and TraceabilityISO – Inspection and Test StatusISO – Control of non-conforming product

GAMP – Good Engineering Practice

ISO – Management Responsibility

GAMP – Good Engineering Practice

ISO – Quality SystemsISO – Statistical Techniques

GAMP – Training of Supplier StaffGAMP – Good Engineering Practice

ISO – Training

GAMP – Good Engineering Practice

ISO – Management Responsibility

CMM Key Process Area

Requirements Management –Establishing a common understanding between the customer and thesoftware project of the customer’s requirement that will be addressed bythe software project

Software Project Planning –Establishing reasonable plans for performing the software engineeringand for managing the software project

Software Project Tracking and Oversight –Establishing adequate visibility into actual progress so that managementcan take effective actions when the software project’s performancedeviates significantly from the plan.

Software Subcontract Management –To select qualified software subcontractors and manage them effec-tively.

Software Quality Assurance –To provide management with appropriate visibility into the process beingused by the software project and the products being built.

Software Configuration Management –To establish and maintain the integrity of the products of the softwareproject throughout the project’s software lifecycle.

Organization Process Focus –To establish the organizational responsibility for software processactivities that improve the organization’s overall software processcapability.

Organization Process Definition –To develop and maintain a usable set of software process assets thatimprove process performance across the projects and provide a basis forcumulative, long term benefits to the organization.

Training Program –To develop the skills and knowledge of individuals so that they canperform their roles effectively and efficiently.

Integrated Software Management –To integrate the software engineering and management activities into acoherent defined software process that is tailored from the organization’sstandard software process and related process assets, which aredescribed in the Organization Process Definition.

CMM Level2

– Re

peat

able

2 –

Repe

atab

le2

–Re

peat

able

2 –

Repe

atab

le2

– Re

peat

able

2 –

Repe

atab

le3

–De

fined

3 –

Defin

ed3

–De

fined

3 –

Defin

ed

Table A. Comparison between CMM and GAMP/ISO 9001. (Chart continued on page 29.)

Page 65: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

much information available on the SEI Web site about thesedevelopments, they shall not be included in this very briefintroduction.

The CMM sets out the criteria that characterize maturesoftware organizations. These criteria can then be used as ameans of appraising organizations and this can be done intwo different ways.

• A software process assessment can be used to deter-mine the state of the processes in an organization. Thelikely outcome of this assessment will be an action plan toaddress priority areas for improvement.

• A software capability evaluation can be used to deter-mine if a software supplier is likely to be able to fulfill anorganization’s requirements.

Organizations being appraised against the CMM are subjectto a rigorous, formal, and independent review by an accred-ited body that will certify them at the appropriate level. TheSEI has an Appraiser Program that aims to select the highestquality candidates for training and authorization. SEI-au-thorized Lead Appraisers are required to submit a report tothe SEI for each appraisal performed. The SEI strives toensure that there is a uniform level of quality applied by allLead Appraisers. The appraisal method includes:

• a maturity questionnaire, which samples the CMM, to becompleted by the organization being appraised

• analysis, by the appraisers, of the responses to the ques-tionnaire

CMM Key Process Area (continued)

Software Product Engineering –To consistently perform a well-defined engineering process thatintegrates all the software engineering activities to produce correct,consistent software products effectively and efficiently.

Intergroup Coordination –To establish a means for the software engineering group to participateactively with the other engineering groups so the project is better able tosatisfy the customer’s needs effectively and efficiently.

Peer Reviews –To remove defects from the software work products early and effectivelyand to develop a better understanding of the software work products andof the defects that can be prevented.

Quantitative Process Management –To control the process performance of the software project quantita-tively. Software process performance represents the actual resultsachieved from following a software process. The focus is on identifyingspecial causes of variation within a measurably stable process andcorrecting, as appropriate, the circumstances that drove the transientvariation to occur.

Software Quality Management –To develop a quantitative understanding of the quality of the project’ssoftware products and achieve specific quality goals. Software QualityManagement applies a comprehensive measurement program to thesoftware work products described in Software Product Engineering.

Defect Prevention –To identify the causes of defects and prevent them from recurring. Thesoftware project analyses defects, identifies their causes, and changesits defined software process, as described in Integrated SoftwareManagement

Technology Change Management –To identify beneficial new technologies (i.e. tools, methods andprocesses) and transfer them into an orderly manner, as is described inProcess Change Management. The focus of Technology ChangeManagement is on performing innovation efficiently in an ever-changingworld.

Process Change Management –To continually improve the software processes used in the organizationwith the intent of improving software quality, increasing productivity, anddecreasing the cycle time for product development. Process ChangeManagement takes the incremental improvements of Defect Preventionand the innovative improvements of Technology Change Management andmakes them available to the entire organization.

Related GAMP or ISO Requirement (continued)

GAMP – Good Engineering Practice

ISO – Management Responsibility

GAMP – Good Engineering Practice

ISO – Management Responsibility

GAMP – Reviews

ISO – Inspection and TestingISO – Control of Quality Records

ISO – Process ControlISO – Statistical Techniques

ISO – Management ResponsibilityISO – Design ControlISO – Statistical Techniques

ISO – Corrective and Preventative Action

ISO – Process Control

CMM Level3

–De

fined

3 –

Defin

ed3

–De

fined

4 –

Man

aged

4 –

Man

aged

5 –

Optim

izin

g5

–Op

timiz

ing

5 –

Optim

izin

g

Table A. Comparison between CMM and GAMP/ISO 9001. (Chart continued from page 28.)

Page 66: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

• a detailed on-site investigation, using the CMM as a guide

• preparation of findings identifying strengths and weak-nesses in terms of key process areas in the CMM

• preparation of a maturity profile

• presentation of appraisal results to organization

Some organizations will be initially assessed at Level 1 or 2and work their way from there, through each level in turn.Others may start with an assessment at perhaps Level 4 andmake improvements over time to gain Level 5. It must bestressed that each level is a building block upon which theprocesses for the next level will be built. And, so even if anorganization is first assessed at a high level, they will almostcertainly have gone through the lower levels in their evolu-tion. Achieving the higher levels can take a considerableamount of effort, and as implied by the name, a considerableamount of time to reach maturity.

The structure of the model is represented in Figure 1. Thisshows the relationship between the elements of the model.Each Maturity Level (Initial, Repeatable, Defined, Managed,and Optimizing) indicates the capability of the organizationand each contains the Key Process Areas. These Key ProcessAreas are at the heart of the CMM, are designed to achieveone or more Goals, and are organized by Common Features.The Common Features (see below for more detail) addressthe Institutionalization of the processes within the organiza-tion. (This is very important – for an organization to reach thehigher levels of maturity, the processes must be an integralpart of the company culture.) Each Common Feature con-tains Key Practices which are descriptions of activities or theorganizational infrastructure in place for each Common Fea-ture.

The Common Features (addressing how the key processesare institutionalized) are:

• Commitment to Perform - describing what the organi-zation should do to ensure the process is established, e.g.setting policies and ensuring that leadership is in place.

• Ability to Perform - describing the necessary precondi-tions to allow the process to become effective, e.g. havingin place the appropriate resources, organizational struc-tures, and training programs.

• Activities Performed - describing the activities, roles,and procedures required to implement the Key ProcessArea, e.g., planning the work, preparing procedures for thework, carrying out the work, tracking progress, and takingnecessary corrective action.

• Measurement and Analysis - describing the basic mea-surement practices carried out to determine the status ofthe process that can then be used to control and improvethe process.

• Verifying Implementation - describing what should bedone to ensure that the activities comply with the estab-lished process, e.g., reviews and audits by managementand quality assurance.

At the heart of the model are the Key Process Areas whichidentify the issues that must be addressed to achieve amaturity level. The Key Process Areas are very briefly de-scribed in Table A.

Level 1 - InitialThe software process is characterized as ad hoc, and occasion-ally even chaotic. There is not a stable environment forsoftware development and during a crisis, the plans andprocedures are likely to be abandoned and all efforts will befocused on coding and testing. Few processes are defined, andsuccess can happen depending on individual effort and hero-ics.

Level 2 - RepeatableBasic project management processes are established to trackcost, schedule, and functionality. The necessary process dis-cipline is in place to repeat earlier successes on projects withsimilar applications. There are likely to be differences inapproach taken by different projects; however, the projectswill be guided by organizational policies. Key Process Areasare:

• requirements management• software project planning• software project tracking and oversight• software subcontract management• software quality assurance• software configuration management

Level 3 - DefinedThe software process for both management and engineeringactivities is documented, standardized, and integrated into astandard software process for the organization. All projectsuse an approved, tailored version of the organization’s stan-dard software process for developing and maintaining soft-ware. Responsibility for the software process activities isassigned and an organization-wide training program willensure that staff and managers have the knowledge andskills required to fulfill their roles. The capability of Level 3organizations is standard and consistent because both engi-neering and management activities are stable and repeat-able. Cost, schedule, and functionality are under control andquality is tracked. Key Process Areas are:

• organization process focus• organization process definition• training program• integrated software management• software product engineering• intergroup coordination• peer reviews

Page 67: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

Level 4 - ManagedProductivity and quality data are collected and analyzed for allimportant software process activities across all projects as partof an organizational measurement program. Projects achievecontrol over their products and processes by narrowing thevariation in performance to fall within acceptable quantitativeboundaries. Meaningful variations can be distinguished fromrandom variation and the risks involved in introducing changeare known and carefully managed. Causes of variation areidentified and addressed. Key Process Areas are:

• quantitative process management• software quality management

Level 5 - OptimizingThe entire organization is focused on continuous improve-ment. It will identify weaknesses and proactively strengthenprocesses with the goal of preventing the occurrences ofdefects. Innovations that exploit the best software engineer-ing practices are identified and implemented across theorganization. Defects are analyzed to determine and elimi-nate root causes. Lessons learned are disseminated through-out the organization. There will be organized efforts toremove waste and inefficiency. Key Process Areas are:

• defect prevention• technology change management• process change management

CMM BenefitsOne of the main published texts on the Capability MaturityModel includes details of a number of organizations and theadvantages they accrued from the implementation of a CMMapproach to improving their software engineering processes.The text includes the following as stated benefits:

• return on Investment (ROI) - one company has reported areturn of 4.5 to 1 in moving from Level 2 to Level 3. Anothercompany reported an ROI of 6.35 to 1

• reduction in defects - customer reported defects from 25%to 10%

• increased productivity - projects on schedule from 51% to94%, schedule slippage from 50% to 1%

• more work - more contracts awarded and more time tocarry out work because of reduced rework time (reworkcosts reduced from 41% to 11%)

The text also lists the following less tangible benefits re-ported by organizations:

• improved employee morale• improved quality of work life• fewer overtime hours• more stable work environment

• lower staff turnover• improved communication• improved quality as reported by customers

All of the above should lead to improved customer satisfac-tion. The Capability Maturity Model is not the only way toachieve this level of customer satisfaction, but it is anotherway that may benefit our industry if it were to be more widelyadopted internally or if we contracted more suppliers whohave adopted the approach.5

The Capability Maturity Model is a complex and ex-tremely lengthy model (it runs to several hundred pages,whereas ISO 9001: 1994 is contained in under 20 pages) thatcan only be very briefly described in an article of this nature;however, it is hoped that this summary will serve as a usefulintroduction for any reader who is unfamiliar with the modeland its uses.

ComparisonsJust as there have been comparisons drawn between theGAMP Guide and ISO 9001, there have been several compari-sons drawn between CMM and ISO 9001 (some of these canbe found on the SEI Web site), and it is widely believed thatan organization at Level 2 of CMM would benefit fromachieving ISO 9001, whereas an organization at Level 3would be likely to have all of the necessary processes in placeto achieve ISO 9001. It is reasonable then to assume thatorganizations achieving Levels 4 or 5 would have processes inplace that are far more rigorous, and effective in ensuringdelivery of consistent quality, than organizations doing onlythat necessary to gain ISO 9001.

It is worth noting here that there will be clear areas ofdifference between the GAMP Guide and CMM. For example,the GAMP Guide covers keeping a computer system in itsvalidated state, post-implementation, and through its usefullife. CMM does not cover the post-delivery elements in greatdetail because its focus is very much on the software engi-neering processes.

The objective of this article is to look at how IT organiza-tions, which have achieved the higher CMM Levels (4 and 5),could be potential suppliers to the industry. And, the ques-tion that this article must answer is: How do the processesneeded to achieve CMM Level 4 or 5 match with the processesneeded to build a system that can be easily validated whenused in the pharmaceutical industry?

Table A provides the CMM levels and the key process areasfor each level. The table structure is based on CMM rather thanGAMP. This is intended to give readers, who are not familiarwith CMM, an insight into its requirements and objectives.

It is worth noting here that a key focus of CMM isrevisiting, reviewing, and refining the processes. So while itmay appear that all of the basic IT processes are in place bythe time an organization reaches Levels 2 or 3, the activityrequired to reach Levels 4 and 5 will mean that theseprocesses are more effective, more efficient, and products ofa better and more consistent quality are being delivered tothe customer.

Page 68: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

8 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Key Points of the ComparisonThe following is a comparison based on the Key Process Areasassociated with each maturity level. The comparison starts atLevel 2 because one of the characteristics of a company atLevel 1 is that it has no defined key processes.

Level 2 - RepeatableAt Level 2, organizations must be able to effectively:

• manage requirements• plan projects• track project progress• manage subcontractors• plan and manage quality assurance activities• plan and manage software configuration management

activities

They will be able to repeat successes when faced with similarpieces of work. This level of maturity compares with organi-zations who have addressed the following GAMP and/or ISO9001 requirements:

• quality and project planning• reviews• good engineering practice• management responsibility• quality systems• process control• corrective and preventative action• internal quality audits

Level 3 - DefinedAt Level 3, organizations will have formal, documented,standardized, and integrated processes for software engi-neering. These will cover the following areas:

• process improvement initiatives at organizational levels

• data gathering for analysis in support of the improvementinitiatives

• planned and managed training activities

• integrated software management – a refinement of projectplanning and project tracking being done at Level 2

• software engineering processes that ensure consistentproduct delivery

• intergroup coordination ensuring that all interactions arecontrolled and effective

• peer reviews leading to defect identification and removal

This level of maturity compares with organizations who havemet all of the previously stated requirements of GAMP and/or ISO 9001, plus the following:

• statistical techniques• training• training of supplier staff• inspection and testing• control of quality records

At Level 3, CMM starts to diverge from both GAMP and ISO9001. This divergence is related to the focus of the approaches– CMM is entirely focused on the organization’s processeswhereas GAMP and ISO 9001 tend to focus (at least in part)on the product of the processes. This process focus arguablywill lead to a much more consistent delivery capability thana product focus.

Level 4 - ManagedAt Level 4, an organization’s processes and products will bequantitatively understood and controlled. The processes re-quired to achieve Level 3 will be built on in the following areas:

• quantitative process management - within a measurablystable process, causes of variation are identified andcorrected

• software quality management - a comprehensive mea-surement program setting goals for software products

It is worth noting here that while the underlying objective ofthe various approaches is similar, the focus of both GAMPand ISO differs from CMM. ISO 9001 Process Control andDesign Control aim to define and document processes so thatthey can be verified and appropriate controls can be imple-mented. While this verification will involve quantitativestatistical and elements, the CMM aims to ensure thatprocesses are measurably stable allowing transient varia-tions to be identified and dealt with.

It is quite likely that the ISO and GAMP requirements forSoftware Quality would be met by a CMM Level 2 companywith their approach to Quality Assurance. However, ISO9001 can be interpreted as requiring some of the elementsassociated with Quality Management implemented by CMMLevel 4 organizations.

Level 5 - OptimizingAt Level 5, organizations will have fully embraced continu-ous process improvement and they will be piloting innovativeideas and technologies. The processes required to achieveLevel 4 will be enhanced in the following areas:

• Defect Prevention - identifying, analyzing, and eliminat-ing the root cause of defects.

• Technology Change Management - proactively seekinginnovation and improvement opportunities and imple-menting them in a controlled manner.

Page 69: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Quality Systems

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 9©Copyright ISPE 2003

• Process Change Management - taking the improvementsidentified through defect prevention and technology im-provements and making them available throughout theorganization.

Organizations who have met the requirements of GAMP and/or ISO 9001 will have addressed Defect Prevention. However,the approach will probably have been reactive and may wellhave been based on customer complaints. The focus of a CMMLevel 5 company will be on root cause analysis and eradica-tion.

ConclusionSo, how do the processes needed to achieve CMM Level 4 or5 match with the processes needed to build a system that canbe easily validated when supplied to the pharmaceuticalindustry?

The comparison summarized above and detailed in TableA shows that there are links from nearly all elements of CMMto the GAMP Guide (or to ISO9001). It also shows that theonly element of ISO9001 not covered in some way in CMM isthat of Servicing. Because of CMM’s focus on the develop-ment processes, the ongoing support and maintenance ofsystems is not addressed in any great detail. As alreadymentioned, there are areas covered in GAMP that are notcovered at all in CMM. However, these are related to specificvalidation activities (and therefore not the responsibility ofthe supplier), to hardware or infrastructure issues (andtherefore outside of the scope of CMM and Software Engi-neering), or to on-going operational issues (again outside ofthe scope of CMM).

The basic premise of both CMM and GAMP, and thegreatest similarity between the two (and this applies also toISO) is that organizations should have defined and docu-mented processes in place and their practices should matchthese – “say what you do and do what you say.” It is reason-able then to conclude that organizations certified at CMMLevel 4 or 5 will be doing all that is required to satisfy anISO9001 audit or indeed a GAMP-style Supplier Audit.

But they will be doing more. They will be constantlylooking at their processes, refining them, and looking foropportunities for improvement. They will be constantly gath-ering data relating to their processes and products andanalyzing that data looking for the root causes of problems sothat they can be eliminated. And, it is very likely that theywill be looking for opportunities to demonstrate to existingand potential customers that this emphasis on quality andimprovement is beneficial to all concerned.

In a final conclusion, I would heartily recommend thatCMM Level 4 or 5 companies be included as potential suppli-ers to any pharmaceutical company looking for IT suppliersof good quality software products and software developmentprojects.

References1. Software Engineering Institute Web site -

www.sei.cmu.edu/cmm. In particular, the following twoarticles have provided very valuable information:

• Paulk, Mark C., Curtis, Bill, Chrissis, Mary Beth, andWebber, Charles V., “Capability Maturity Model forSoftware, Version 1.1” Technical Report CMU/SEI-93-TR-24, Software Engineering Institute, Carnegie MellonUniversity, Pittsburgh, USA, February 1993.

• Paulk, Mark C., “How ISO 9001 Compares with theCMM,” IEEE Software, January 1995.

2. The information on the GAMP Guide was gleaned fromversions 3 and 4 of the guide.

3. BS EN ISO 9001: 1994, Quality Systems, Model for Qual-ity Assurance in Design, Development, Production, Instal-lation, and Servicing, BSI 1994.

4. FDA Guidelines on General Principles of Process Valida-tion, May 1987.

5. Carnegie Mellon University, Software Engineering Insti-tute, Paulk, Mark C., Webber, Charles V., Curtis, Bill,Chrissis, Mary Beth, “The Capability Maturity Model:Guidelines for Improving the Software Process,” AddisonWesley Longman Inc, 1994.

AcknowledgementsThanks are due to the people who reviewed early drafts of thisarticle – they have a greater and more detailed knowledge ofeither CMM or GAMP than I do and they have had thestamina to get through it and provide me with feedback.

Colin Jones, ConformityRoland Nicholas, IT SolutionsSrikanth Rao, IT SolutionsDavid Selby, Selby Hope InternationalSteve Watson, IT Solutions

About the AuthorLorna Foote is a Director of Ancrum Asso-ciates Ltd., specializing in the provision ofconsultancy in IT Quality, Regulatory Com-pliance, and Validation. Foote has 25 yearsof IT experience, including 15 years in phar-maceutical manufacturing and three yearsin finance in the city of London. She hasspecialized in leading teams, building pro-

cesses, and delivering products that achieve regulatory com-pliance, satisfy internal and external auditors, and deliverresults of excellent quality. Foote has developed and imple-mented Quality Management Systems for a number of differ-ent IT departments, is an ISO 9001 auditor, and contributedto an early version of the GAMP Guide. At the time of startingthis article, Foote was employed by IT Solutions (Europe)Limited, a branch of ITS Inc. whose development centers inBangalore and Chennai in India are both CMM Level 5certified. E-mail: [email protected]

Page 70: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1©Copyright ISPE 2003

Production of Purified Water in theBiotechnology Industry

by Patrick Chkroun, Denis Acker, and Philippe Jaunin

This articledescribes acontinuous,five-phasetreatmentsystemproducingpurified waterto USP XXIV.

Biotechnologyin the Service of Health

Today, three main methods exist to pro-duce medicines:

• extraction and purification of natural sub-stances

• chemical synthesis• biotechnology

In healthcare, modern biotechnology utilizingDNA recombination techniques has improvedprevention, diagnosis, and treatment of nu-merous illnesses since the late 1970s.

Since the middle of the last century, Seronohas been producing medical products by tradi-tional methods of extraction and purification ofnatural substances. They have been using the

recombinant DNA methods for more than 10years now. The first “recombinant” product, thegrowth hormone, is being distributed today inmore than 50 countries.

State-of-the-Art TechnologiesThe new biotechnology laboratories in Corsier-sur-Vevey, Switzerland, are among the mostmodern worldwide from the technological view-point, which constitutes a new approach topharmaceutical production based upon cellculture and development. Taking into consid-eration the specific and very complex charac-teristics of the equipment, Serono went intopartnership with architects, engineers, plan-ners, and constructors, who are experiencedand have a good track record in project engi-neering of pharmaceutical facilities, and morespecifically, in biotechnology.

Figure 1. Diagram ofproduction anddistribution of purifiedwater.

Reprinted from The Official Journal of ISPE

PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

Page 71: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

2 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Basic ConceptThe process engineering design concept was conceived by anengineering office in conjunction with the engineering de-partment of Serono in Geneva. The following equipment andservices were supplied:

• purified water production• injection control and destruction of ozone• service water softening (cooling towers steam)• control panels - switching and control• engineering of material and programming conception• commissioning• validation DQ and IQ• assistance in validation OQ

Project GoalsStarting with potable water distributed by the Service desEaux de Vevey (municipal water supply), the aim was toproduce purified water according to the US PharmacopoeiaUSP XXIV, the site specifications of Serono, and to conformto the concepts of installation, manufacturing, and validationmandated by the cGMP. The purified water will be used tofeed the following:

• Multistage Still• Purified Steam Generator• Pre-Washing System• Steam Boiler (mainly with softened water).

In the first stage, the production capacity for purified wateris 6.5 m3/h at 15°C. To guarantee the nominal capacity overall seasons, the system was dimensioned for a water tempera-ture of 10°C - Table A.

Description of thePurified Water Production Process

The installed water treatment system consists of five phases:

• Pre-treatment with multimedia filtration, softening,dechlorination, feed tank with UV-radiation and heatexchangers. These elements are sized for the require-ments of the final extension.

Table A. Physico-chemical parameters required by QA.

Units Value

Aspect/Color clear/colorless

Smell/Taste odorless/tasteless

Conductivity (25°C) µS/cm < 1.25

TOC mg/l < 500

Aerobic microbial contamination CFU/100 ml < 10

Pathogenic micro-organisms

E. coli, enterococci, n/100 ml n.d.pseudomonas aeruginosa,pseudomonas cepacia

Endotoxin content EU/ml < 0.25

Figure 2. Multimedia filtration.

• Demineralization by two-stage reverse osmosis includingneutralization of carbon dioxide. These units are laid outto allow for future extensions.

• Hot water sanitization.

• Purified water storage tank with distribution and circula-tion equipment.

• Disinfection of the treated permeate and of the purifiedwater storage tank by electrolytically produced ozone;ozone destruction by UV-radiation in the feed of thedistribution loop.

Process Description of thePurified Water Production Plant

Pre-Treatment - Multimedia FiltrationSuspended matter in the raw water is removed by passing thewater through two multimedia filters in parallel. Each unitis filled with quartz sand and activated carbon, and has acapacity of 36 m3/hr. The system is configured to work inparallel. During backwashing a single unit works.

The control of each unit is independent, allowing each unitindependent response to differential pressure and feed watersolids capacity. The signal to backwash a unit can be given byone of three parameters: differential pressure, water con-sumption, or cycle time. Differential pressure, which indi-cates that the filter is becoming saturated, has priority overcycle time in initiating a backwash. Starting the backwashprocedure requires an operator to activate the automaticcounter-current washing process at the control panel. Thedifferent steps of this operation can be followed - Figure 1.

Page 72: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3©Copyright ISPE 2003

SofteningThe exchange of calcium and magnesium ions with sodiumions takes place in a series of softeners filled with ionexchange resins specially developed for potable water andfood applications. The unit allows softening with both col-umns either in series or in parallel. Each unit is equippedwith the necessary instrumentation to measure differentialpressure, water consumption, and flow. In the softened watercollector, a resin trap retains the resin fines and any resinsthat might have accidentally come from the softeners. Theresin retention is indicated by the pressure loss measure-ment installed with the trap. A valve arrangement allowspurging of the filter on request. Three parameters of eachsoftener are permanently controlled: differential pressure,softened water production, and cycle time. When the pro-grammed capacity is reached, the working column is regen-erated. The polishing column automatically becomes theworking column. At the end of the regeneration cycle, theregenerated column becomes the polishing filter. The re-sidual hardness is constantly monitored with an alarm levelof 1 mg/l. To guarantee this value monodisperse resinsspecially developed for potable water and food applicationsare used, regenerated co-currently with 90 g NaCl per liter ofresin. Concentrated brine, stored outside of the building isused for regeneration. A pump transfers the brine, controlledby level sensors, to the day tank located in the technical zone.

The regenerations are interlocked so that softener regen-eration and multimedia backwash cannot occur simulta-neously. Softener regeneration has priority - Figure 2.

DechlorinationThe possible presence of free chlorine in the feed water isdetected by continuously measuring the Redox potential. Ifchlorine traces are present, a signal interrupts the softenedwater feed. This can only be reset into operation once bi-sulfite dosing has been actuated and the measured chlorinelevel is back within control limits. A second Redox measure-ment installed right at the feed of the reverse osmosis unitensures security by stopping reverse osmosis productionwhen chlorine is present. In this case, the softened water isrecirculated into the feed tank.

Feed TankThis tank receives three flows:

• softened feed water• recirculating permeate• concentrate during recirculation or for out of specification

returns (Redox - pH)

The softened water, possibly dechlorinated, is automaticallyfed into the tank via two “spray-balls” depending on the waterlevel. The measurements of level, service pressure, andtemperature are linked to the control screens. The reverseosmosis is fed by a group of variable speed electropumps,pump rate being dependent on whether the reverse osmosisor the recirculation is being fed. The tank vent is equipped

with a sterile microfilter in a heated housing to eliminatecondensation and with a carbon dioxide adsorber in theoutlet. Connections with the sterile filter allow in-line integ-rity tests and validation of the filter element. This equipmentlimits pH-variations in the feed water.

This very specific layout was chosen to continuously keepthe feed network for both the reverse osmosis unit and theproduction of purified water in a dynamic circuit with theobjective to avoid water stagnation and to limit as far aspossible the loss of water in concentrate or rinsing water.

UV-SanitizationDownstream of the feed pumps, a UV-sanitizer at 254 nmwith an intensity of more than 30,000 microWatts/cm2, en-sures reduction of bacterial contamination in the reverseosmosis feed. The radiation intensity of the lamps, the oper-ating status, and the alarms are given to the control screen.

Heat ExchangersThe feed of the reverse osmosis is equipped with two tubularheat exchangers with double-plates, water/steam and water/cooling water, of sanitary design, installed in series.

In manual operation, the feed temperature of the reverseosmosis unit is kept steady at 15°C.

Reverse OsmosisIn the final installation, two lines of reverse osmosis units,each with a capacity of 13.5 m3/h, will be in operation. Atpresent, one unit is installed with a capacity of 6.5 m3/h,which may be extended to 13.5 m3/h. Any suspended residueis retained by microfilter cartridges with a porosity of 1.2micron.

The pH of the feed water is continuously controlled toneutralize and transform carbonic acid into bicarbonate.Caustic soda, pharmaceutical grade - is only injected whenthe system is not in recirculation.

The double-stage reverse osmosis unit is fed by only onepump. The permeate and concentrate capacities are con-trolled by the panel as well as all parameters of pressure andtemperature. The panel stops the high-pressure pump whenit reaches the pressure and temperature limit values - Figure3.

Permeate capacity @ 15°C:expected reached

1st stage m3/h 6.5 6.52nd stage m3/h 13.5 13.53rd stage m3/h 25 25

SanitizationThe water/steam exchanger produces hot water at 80°C forthe sanitization of the feed tank, the feed pumps, the UV-generator, the security microfilters, the reverse osmosis feed,and the recirculation piping. During this operation, themembrane stages are by-passed by a “spool piece.” The samesystem is used for the sanitization at 80°C of the pretreat-ment, that is the multimedia filters and the softeners. The

Page 73: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 4. Double-stage reverse osmosis (extensible).

contents of the feed tank are used for the sanitization itself.It is actuated at the panel; the UV-lamp is switched off, andthe process runs automatically. When the control indicates atemperature of 80°C, circulation is maintained at that tem-perature for one hour. At the end of the cycle, cooling isautomatically initiated by means of the exchanger (water/cooling water) until a temperature of 15°C is reached. Theproduction processor is then directly reset into normal opera-tion by means of the panel. All sanitization phases areautomatically recorded.

Purified Water Storage TankThe permeate mainly feeds the stainless steel 316 L tankthrough an automatic diaphragm valve depending on thelevel indicated by the differential pressure measurement.The double-walled tank - for cooling and maintaining thepurified water temperature at 18°C - is equipped with thefollowing elements for reasons of security and a sterileatmosphere:

• bursting disk in case of excess pressure with a controlcontact to an open air outlet

• continuous pressure and temperature control• security controls for high and low levels• sterile microfiltration vent filter kept at 80°C (to avoid

condensation) with connections to allow in-line testingand validation

• CO2-adsorber, installed upstream of the vent filter allowsair passage over a bed of chemically pure calcium gran-ules. The adsorption capacity of these granules is 1 mol ofcalcium hydroxide neutralizes 1 mol of carbon dioxide.

The circulation return is split into two streams. One of themfeeds the water with a high ozone concentration and a

capacity of approximately 4.7 m3/h into the tank beneath theminimum water level. The other one feeds the main returninto the tank by three spray balls at a pressure of 2 bar.

Disinfecting by OzoneThis system was chosen to allow a continuous disinfection ofthe purified water in circulation, in the storage tank, and ofadditional permeate from the double-stage reverse osmosis.Furthermore, it allows sanitization of the entire distributionnetwork without additives, without modifying the purifiedwater quality, and without loss of rinsing water. Ozone is atri-atomic modification of oxygen. It’s an extremely strongand effective oxidant. The high reactivity allows effectivedestruction of microorganisms and guarantees continuoussanitization. This strong oxidant is produced by catalyticseparation of water inside an electrolytic cell composed of ananode, a cathode, and a solid polymeric membrane which actsas an electrolyte and separator between the two half-cells.The Direct Current (DC), which is fed to the cell, causes thesplitting of the passing demineralized water into ozone,oxygen, and protons at the anodic side and the reduction tohydrogen gas at the cathodic side. The high-purity ozone thusproduced within the flow-electrolysis cell is immediatelydissolved in the passing purified water stream. This systemworks for demineralized water with a maximum conductivityof 20 µS/cm.

The disinfection system is composed of three groups ofelectrolytic ozone generators fed from the main loop return.The intensity is controlled to keep an ozone concentration ofapproximately 0.1 to 0.2 ppm in the purified water tank. Theozone content is measured at five points in the loop bymicroprocessor analyzers with a measuring range of 1 ppb.All values are given to the panel together with alarm signals(outlets). The polarographic measuring probe is composed ofthree precious metal electrodes, immersed in an electrolyte

Figure 3. Softeners in series.

Page 74: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5©Copyright ISPE 2003

solution, separated from the liquid by a gas-permeable mem-brane.

A protective electrode enveloping the measuring electrodeblocks the influence of other gases and improves the stability.A voltage difference is applied between the electrodes toreduce the ozone passing the membrane thanks to a partialpressure gradient. A proportional electric current is gener-ated, measured by the analyzer, then calibrated, displayed,and converted. The value is available in analog and numericoutputs - Figure 4.

Piping and Purified Water DistributionTwo centrifugal pumps in stainless steel 316L each with acapacity of maximum 24.9 m3/h with open wheels and speed-variable motors ensure the purified water distribution. Thecapacity is set by means of a pressure detector. They can beindependently isolated from one another by an automaticdiaphragm valve arrangement controlled by the panel. Theozone is destroyed by two UV-generators in series working atan intensity of above 90,000 microWatts/cm2, and in theposition to treat a maximum flow of 49.8 m3/h.

All equipment parts in contact with the purified water aremade of electropolished stainless steel 316L. All controlparameters – intensity, temperature, and operating status -are transmitted to the control panel - Figure 5.

On the fittings and valves of the multimedia filters, thesofteners and the reverse osmosis plants, the straight valvesare placed in strict compliance to the 3D guidelines in orderto reduce the dead ends in all of the fittings to a minimum.Starting with the softening station, all valves are membranevalves made of materials complying with the US FDA andwith housings made of wrought stainless steel 1.4435. Thevalves connected on a bypass as well as the sampling valvesand the taps are T-valves (zero dead-leg body) of identicalquality. The valve weir placed immediately at the point of

Figure 5. Ozonization plant (3 x 4 gO3/h).

Figure 6. UV generator for the ozone destruction.

flow (straight throughflow enables almost complete deadleg-free take). The material of the pipes is specified by theengineering office and is adapted to the conveyed liquids. Forthe stainless steel hygienics tubing and fittings materialgrade 1.4435 is used with low ferrite, fully annealed (< 0.5 %).The interval surface has a polished scratch- and pit-freeroughness not exceeding 0.6 µm Ra. For the purified section,pipes, instrumentation, and valves are assembled by orbitalwelding without insertions with a ferrite content < 1 % or byusing BBS connections. An endoscopic control has been ef-fected for each welding and the ferrite content measured. Tenpercent of the weldings and all manual weldings (approxi-mately 3 % of all weldings) have been controlled with X-rays(Rx). The entire weldings are documented.

Sanitization of thePurified Water Distribution System

The tank and its equipment as well as the distribution systemmay be sterilized by purified steam or ozone. The usualprocess is a periodical sanitization at night, or when stipu-lated by the user. During this operation, the panel automati-cally interrupts all feeds to consumers and the UV-lamps.The ozonated purified water circulates within the wholedistribution loop for a preset period to obtain a constantoptimum ozone concentration in the circulation return. Dueto the length of the distribution loop, it takes 17 minutes toobtain an ozone value in the return. Then, the UV-systemsare returned to operation. The end of the sanitization ismarked by the absence of ozone in the output of the secondUV-generator. The panel can control and vary all proceduredurations. Sanitization by purified steam is used only inextreme cases, for example, after opening the loop for main-tenance, piping modifications or replacing elements. Thepurified steam is introduced by a flexible connection into thepermeate line directly downstream of the reverse osmosis

Page 75: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

6 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

Figure 7. Operating and control panel, Dosing group for causticsoda and sodium bi-sulfite, Double-stage reverse osmosis(extensible).

unit. The manual valves by-pass the ozone generators atshared in- and outputs. All purge valves as well as the ventingvalve of the sterile microfilter are opened and the pressure ofthe purified steam is set at 2.5 bar. The sanitization processis then actuated and entirely controlled by the panel. Allcondensates are recovered and drained. When all measuringpoints indicate the sanitization temperature (121°C), this issteadily maintained over 30 minutes. Then, after stoppingthe steam, sterile process air is fed into the sterile microfilterin counter-current. Simultaneously, cooling water circulatesbetween the double jackets of the purified water storage tank.The cooling operation is stopped when the purified watertemperature reaches 18°C in the return. The normal distri-bution process is then reset into operation.

System ManagementAll functions are managed by a programmable logic control-ler situated immediately next to the installations. The dis-play and touchscreen allow supervision of the entire purifiedwater production system and also allows modification ofcertain parameters when needed. The link to the controlcenter is where all data is recorded via a netware system.Data may be reproduced in different ways depending on theinformation priority and the hierarchic treatment - Figure 6.

Description of the Service Water TreatmentThe service waters comprise the feeds for the following:

• Cooling Towers• Supplement to the Steam Production• Compressor Cooling

The raw water is directly fed to a series of softeners workingalternately. The same specifications as those applied for thesoftening unit for the “purified water production” are used.

Steam Boiler FeedThe steam boilers are fed first by the condensate return,secondly permeate, and then finally, softened water. If per-meate is required for the purified water storage tank, anautomatic valve manifold switches the feed to softened watercoming from the softening unit. This unit has its own controlcabinet with programmable logic controller and touchscreen.All information is also transmitted to the control center.

Qualification and ValidationWhy Validate?Only by process validation can you ensure that all units meetthe required quality standards; quality control alone is notsufficient to guarantee that the product quality is achieved.These procedures reduce costs by reducing expensive prob-lem solving. The concept of defining the basis is regulated byseveral organizations: no matter which organization is fur-ther mentioned, validation is compulsory.

Three terms are often used for this concept, the definitionsof which are as follows:

• Calibration - all operations that establish under certaingiven conditions the relation between values indicated bya measuring apparatus or system or the values given by anequipment measurement and the corresponding values ofa standard measure.

• Qualification - operation with the aim to demonstrate thatequipment operates correctly and in fact provides theexpected results.

• Validation - Establishment of the proof in conformity withthe principles of Good Manufacturing Practice (GMP) thatthe execution and the use of all processes, procedures,materials, raw materials, products, activities, and sys-tems effectively ensure reaching the expected results.

ValidationInstallation Qualification - IQAim: to identify each piece of equipment and to determinewhether it is installed in conformity with the approvedspecifications.• System Description• Utility Requirements (electric capacity, cooling water, air

steam, nitrogen drainage)• Specifications of the Different Plant Components• Calibration of the Measuring Instruments• Training and Qualification respectively of the Operators• Adequate Environment

The installation qualification must demonstrate the follow-ing:

Page 76: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 7©Copyright ISPE 2003

• the system was laid-out and built in conformity with thespecification

• the equipment performances are within specification andwere tested by the manufacturer

• the components are installed correctly and the utilities arein line with the equipment requirements

• the operators are of the technical standard required for thecorrect operation and maintenance of the plant

• the equipment control and monitoring instruments areprecise

• the environment is of the required quality

All these operations aim to show us that the system and itsenvironment meet the specifications.

Operational Qualification - OQAim: to determine whether the plant functioning conforms tothe approved specifications. The objective of this stage is toensure that the tested system performances are adapted tothe process for which the system is intended.

One will thus:

• test the equipment performances• test the cycles• test the programs

All these operations aim to prove that the system in isolationfunctions according to specifications.

Performance Qualification - PQAim: to test the equipment functioning in its normal capacityrange over one year. Still called “performance qualification”by the Americans, this operation whether prospective, retro-spective, or consecutive must give proof that the system inproduction conditions, allowing for seasonal variations, givesthe expected results.

All these operations aim to show that the whole systemperforms to specification in production conditions and at alltimes.

DocumentationGood documentation is one of the essential and indispensableelements of the system quality assurance.

Functions:• To give instructions and working procedures:

- uniformity of intervention maintenance and utilizationpractices

- adherence to regulations- adherence to validation files- avoidance of deviations- reference bases

• Recording of information:- historic- traceability- evidence

Elements constituting the documentation system:• specification of plant components• specification• general procedures• reports of calibration, qualification, and validation• material follow-up• diagrams, drawings (P&ID), and programming listings• files of malfunctions, anomalies, and change control pro-

tocols• recordings of maintenance activities• recordings of certain material controls and certifications

LabelingThe documents are labeled according to a pre-establishedsystem allowing their management. The labeling proceduremay comprise the following headings:

• Service Symbol• Nature of the Document Symbol• Field of Application Symbol• Edition Number• Version

ProductionThe production of the document is carried out by a qualifiedperson, the closest possible to the user.

VerificationThe newly written or revised document may be verified byany qualified person (in direct contact with the user of thedocument).

ApprovalAny document must be approved by qualified persons. Theirrole is to make the documents compulsory and official.

DistributionEnsured by the Quality Assurance Department, it makesadequate numbers of copies available to all users.ArchivingThe circulating copies are destroyed, the original archived.

The ProceduresDocuments that give instructions to allow a trained person toperform an operation in conformity with an established rulein a way that the expected result can be reached in a repro-ducible manner.

ConclusionThe plant has worked continuously since its start-up (end of1998) without any interruption other than the security stopfor passing into the year 2000. All operating parameters,

Page 77: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Purified Water

8 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 2003

daily controls by the user are in line with the expected valuesas far as quantity is concerned and always superior as far asquality is concerned.

About the AuthorsPatrick Chkroun is a Technical ServicesManager at Laboratories Serono SA inAubonne, Switzerland. His interests focus onthe technical requirements of equipment andinstallations dedicated to the pharmaceuti-cal industry. Having graduated in chemicalengineering from the Institute of Sciences ofRouen, France, he also holds an MS in physi-

cal organic chemistry from the Univeristy of Paris. He has 10years of experience in engineering and maintenance in sev-eral pharmaceutical companies.

Laboratoires Serono S.A., Z.I. de l’Ouriettaz, CH-1170Aubonne VD, Switzerland, tel: +41-21-821-74-33, email:[email protected].

Denis Acker is the Clean Systems Respon-sible at Laboratories Serono SA in Corsier-sur-Vevey, Switzerland. He obtained hisFrench university degree in material phys-ics, as well as a Swiss technical degree inthermal physics. He also has wide experi-ence in physical assays and analyses, valida-tion, and metrology. He has been responsible

for clean systems for the past eight years within the SeronoLaboratories.

Laboratoires Serono SA, Z.I.B., CH-1809, Corsier-sur-Vevey, Switzerland, tel: +41-21-923-23-53 email:[email protected].

Philippe Jaunin holds a Bachelor degree inchemical engineering and is working in thedepartment serving the pharmaceutical andlife science industry of Christ Ltd. He hasbeen working in water treatment for 30 years.

Christ SA, Subsidiary of Puidoux, Z.A. LeVerney, CH-1070 Puidoux, Switzerland, tel:+41-21-946-34-42, email: [email protected].

Page 78: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Barrier Isolation

This article Reprinted from The Official Journal of ISPE

summarizes PHARMACEUTICAL ENGINEERING® March/April 2003, Vol. 23 No. 2

survey results taken in 2002 on the use of barrier isolators in fill/finish applications in the parenteral industry.

For complete survey raw data, click here.

Figure 1. Barrier isolator filling line - deliveries by year.

Barrier Isolation History and Trendsby Jack Lysfjord and Michael Porter

This is the third survey (1998 and 2000 ceuticals with the Barrier Users Group Sympo­are previous surveys) on the use of sium (BUGS) and LUMS groups (Lilly, Upjohn, Barrier Isolators for automated phar- Merck, et. al.). However, there was always a maceutical fill finish operations that benchmarking question as to usage in the in-

was implemented. Manual operations in a glove dustry. A decision was made to try and survey box are not considered. We began this journey users and equipment manufacturers to get in the late 1980s and early 1990s when the use some answers. Keep in mind that the data is of pharmaceutical “barriers” or “isolators” was only as good as the input from many sources a discussion concept rather than a practice for and that while trends certainly apply, numbers improving sterility assurance. New ground was should not be viewed as absolute, but rather as broken developing some of the first “produc- trends over time. Every fill finish application tion” fill/finish barrier isolators for pharma- for Barrier Isolators may not have been

found, but the vast majority for trend analysis were identified. Preliminary data was presented at ISPE confer­ences in Arlington, VA in June 2002 and in Zurich in September 2002. Here are the results for 2002:

Filling Barrier Isolators Delivered:1998 2000 2002

84 172 199

Figure 1 shows that global deliveries of Barrier Isolators for automated fill fin­ish are continuing from 1996 through 2002 with about 20 per year being delivered.

Figure 2 shows the number of deliv-Figure 2. Barrier isolator eries for various continents. Of note infilling line - deliveries by

2002: European deliveries dropped,year and region. North American deliveries maintained, and Asia (Japan) is taking the lead in more recent application of this technol­ogy.

Figure 3 shows companies with the highest usage. It is interesting to note the commitment some companies put into this type of technology. It is also interesting to note the reluctance of oth­ers to begin the learning process with barrier isolation.

©Copyright ISPE 2003 MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 1

Page 79: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Barrier Isolation

Figure 3. Barrier isolator filling line - companies with highest usage.

Figure 6. Type of container.

Figure 4. Months from delivery to start-up.

Figure 5. Filling line type.

Figure 7. Maximum speed.

The following are the number of companies using barrier isolation:

1998 2000 200238 56 67

The following are the number of units in operation:

1998 2000 200234 70 90

The number of units in operation is close to half of the number of delivered units. The remainder are being installed, com­missioned, validated, and approved.

PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003 ©Copyright ISPE 20032

Page 80: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 3

Barrier Isolation

One of the more interesting pieces of data is the number of filling lines with FDA approval (out of 128 responses):

1998 2000 2002 6 26 52

Figure 4 indicates the number of months from delivery to start-up. Data is wide spread and has a strong correlation to line speed and degree of difficulty in dealing with processing inside the enclosure. Slower speed lines (clinicals) were easier to start-up and vice-versa. Typical clinical applications do not go through the rigorous validation process that higher speed production systems require.

Figure 5 shows “Filling Line Type” by liquid, powder, or both applications.

Vials are the predominant container used with Barrier Isolation Systems as shown in Figure 6.

The maximum speed of Fill Finish Systems is shown in Figure 7. Note: the large number of applications for 0 - 99/min.

Table A shows hard wall construction (stainless steel and glass) predominates due to:

1. Robustness/Reliability 2. Less Absorption of Sterilant 3. Rigidity prevents breathing of Barrier Isolators (creating

negative pressure inside and ingesting particles through openings when removing hands from gloves)

Table B shows surrounding room classification. Class 100,000 (1SO class 8) is the predominant choice. Note that some early applications with unclassified surrounding rooms have been upgraded to class 100,000 (ISO class 8).

Hydrogen Peroxide Vapor is the sterliant of choice as shown in Table C. Use of half-suits is shown in Table D.

Gloves are the typical method for intervention. One or two-piece gloves usage is shown in Table E.

Table F indicates smooth sleeves or gauntlets are pre­ferred by 4:1 over pleated style.

Glove Replacement period is critical to assurance of glove integrity. Typical replacement periods reported are shown in Figure 8. Typical replacement frequency depends on inter­vention frequency, design of the glove/isolator junction and manipulations typically performed at a particular glove loca­tion (particularly when broken glass is present).

Integrity testing and method of testing are shown in Table G and Figure 9, respectively.

Use of a second disposable glove (latex) is shown in Table H. This is typically placed on the hand prior to going into theglove port, but some use it over the glove port inside the isolator.

The use of mouseholes and depyrogenation tunnel open­ings creates an “open isolator.” Typical ratio of open vs. closed applications is shown in Table I.

The internal environment pressure to the surrounding

1998 2000 2002 Soft Wall 9 9 9 Hard Wall Total 82

73 134 166 143 175

Table A. Barrier isolator construction.

1998 2000 2002 100 3 4 5 1,000 3 4 7 10,000 13 30 40 100,000 55 84 105 Unclassified 5 7 Response Total 79

17 139 164

Table B. Barrier isolator surrounding room classification.

H2O2 Vapor H2O2 Spray Miscellaneous Peracetic Acid 6 Alcohol Wipe 3 H2O2 + Steam 1 Formalin 1 ClO2 1 Other 5 Total

135 13

165

Table C. Barrier isolator sterilants.

1998 2000 2002 17 21

*Question Not Asked (102 Responses) (126 Responses)

Table D.The number of lLines with half-suits.

One Piece Two Piece Total 3 11072

Table E. Number of filling lines that use - one or two piece gloves.

Yes No Total 14 56 70

Table F. Two piece glove - is gauntlet pleated?

Yes No Total 91 29 120

Table G. Gloves - do you integrity test?

Yes No Total 78 31 109

Table H. Do you use a second disposable glove?

Open Closed Total 100 30 130

room and pressure with a washer/tunnel are shown in Fig- Table I. Is barrier isolator open or closed?ures 10 and 11, respectively.

©Copyright ISPE 2003 MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 2

Page 81: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

4 PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003

Barrier Isolation

Yes No Total 19 20 39

Table J. Do you use a sterilizable cool zone with depyrogenation tunnel?

Yes Chemical Biological 5 Chemical Biological 5 Chemical Nuclear 2

No Total 10

32 20

76

Table K. Barrier isolators indicating the need for containment.

Figure 8. Glove replacement period.

When a depyrogenation tunnel is used, sterilizable cool zone usage is shown in Table J.

The need for containment is increasing. The need and type of need is shown in Table K. 30% of those reporting indicate some need for containment!

In 2001, Stewart Davenport, Pharmacia, Kalamazoo, MI, reported at the Arlington, VA ISPE Barrier Isolation Tech­nology Conference on media fill data for 3 RABS (Restricted Access Barrier Systems) equipped lines. Data indicated equivalence to isolators for sterility assurance. This year our survey indicates 52 RABS lines delivered. This can be an alternative to isolators when only Asepsis and not containment is needed. This number is in addition to the 199 Barrier Isolators delivered in 2002.

Cumulative deliveries of Barrier Isolators for fill finish are shown in Figure 12. The increase in numbers continues at a fairly consistent rate. An interesting comment from Lennart Ernerot of The Swedish Inspectorate at the 2002 ISPE Zurich Barrier Isolator Conference was that he would mandate the use of isolators for products such as vaccines and other biopharmaceuticals that are slated for children because of

PHARMACEUTICAL ENGINEERING MARCH/APRIL 2003

Figure 9. Method for integrity testing of gloves.

Figure 10. Pressure to surrounding rooms (12.5 Pascals = 0.5” Water).

the known tremendous improvement in sterility assurance over conventional cleanroom aseptic processing. As regula­tors and users of barrier isolators evolve toward this mindset, the slope of the cumulative deliveries line is likely to increase.

Survey Conclusions:

• slight slowing of orders in 2001- 2002

• Europe typically has twice the NA order rate (1995-2001); however, Europe dropped in 2002 to less than NA.

• Asia really accepting technology, particularly Japan (27)

• some companies really accepting technology (Baxter) while others avoid it

• vials predominate applications

©Copyright ISPE 2003 4

Page 82: Reprinted from PHARMACEUTICAL ENGINEERING · Reprinted from PHARMACEUTICAL ENGINEERING ... quality R&D as well as production sites in Japan. As local manufacturers set their sights

Barrier Isolation

• class 100,000 surrounding room • gloves only – minimize half suits • two-piece gloves with smooth sleeves or gauntlets • doing glove integrity tests • using disposable glove (2nd glove)

Remember, you are not only deciding how to produce today’s products, but consider that the decisions you make today will determine what you can do for the next 25-30 years in your facility. Asepsis and containment are the concerns for the future. Will you make the correct choices for your company?

If you have additional information, please contact the authors.

About the Authors Jack Lysfjord is Vice President of Consult­ing for Valicare, a division of Bosch Packag­ing Technology. Since 1981, Lysfjord has held a variety of management positions with Bosch Packaging Technology (formerly TL Systems Corporation). Prior experience was with Dahlberg, Litton, Medtronic, and Onan. Lysfjord is a member of ISPE, recipient of the

2001 Richard Purdy Distinguished Achievement Award, a past member of the Board of Directors, a past Member of the Year in 1994, past Chairman of the Vendor Committee, and past Chairman and current member of the Marketing Advi­sory Council. He holds other memberships in the Parenteral Drug Association (PDA), the American Glove Box Society (AGS), The Parenteral Society (UK), R3 Nordic (Clean Room Processing) the Society of Automotive Engineers (SAE), and the Barrier Users Groups Symposium (BUGS) of which he is the Chairman. He is a frequent speaker and course leader in the US, Europe and Asia, and has been author and co-author for numerous technical papers and articles. Lysfjord holds a BS in mechanical/industrial engineering from the University of Minnesota and an MBA from the University of St. Thomas.

Valicare Division, Bosch Packaging Technology, 8700 Wyoming Ave. N., Brooklyn Park, MN 55445.

Figure 11. Pressure to washer rooms (12.5 Pascals = 0.5” Water).

Figure 12. Barrier isolator filling line - cumulative deliveries.

• usage for lines below 100/minute are most frequent users of isolators

• hard wall Isolators/H2O2 Vapor Sterilants/Class 100,000 Room

• predominate applications

• two-piece gloves without pleats is typical as is second inner glove

Trends for decision makers – benchmarking information for those who are just getting started:

• hard wall isolator • vaporized H2O2 sterilant

Michael E. Porter is a Principal Engineer in the Sterile Process Technology and Engineering group of Merck & Co., Inc. Since 1987, he has held a variety of engineering and supervisory positions within the Merck Manufacturing Divi­sion, focusing on manufacturing, lyophilization, barrier iso­lation technology, filling of vaccines, and sterile pharmaceu­ticals. Porter has prior experience in plant and process design in the petrochemical industry and holds a BS in chemical engineering from Villanova University. He is a member of ISPE and has presented on the subject of cleanroom robotics and barrier isolation technology more than a dozen times to the pharmaceutical community. Porter has been on the steer­ing committee for ISPE’s Barrier Isolation Technology Wash­ington Conference for the past five years.

Merck & Co., Inc., Sumneytown Pike, PO Box 4, WP62C­M10, West Point, PA 19486.

©Copyright ISPE 2003 MARCH/APRIL 2003 PHARMACEUTICAL ENGINEERING 5