44
NOVEMBER 2015 Volume 27 Number 11 Exploring the Potential of Continuous Coating FORMULATION Topical Bioequivalence PEER-REVIEWED Glass Delamination SUPPLY CHAIN Track and Trace

Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

NOVEMBER 2015 Volume 27 Number 11

Exploring the Potential of

Continuous Coating

FORMULATION

Topical Bioequivalence

PEER-REVIEWED

Glass Delamination

SUPPLY CHAIN

Track and Trace

Page 2: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

West seeks partners for its SmartDose electronic wearable injector technology platform. This platform

is intended to be used as an integrated system with drug filling and final assembly completed by

the pharmaceutical/biotechnology company. The SmartDose system, HealthPrize platform

integration is for conceptual purposes only.

West and the diamond logo and By your side for a healthier world™ are registered

trademarks or trademarks of West Pharmaceutical Services, Inc., in the United

States and other jurisdictions. SmartDose® is a registered trademark of Medimop

Medical Projects Ltd., a subsidiary of West Pharmaceutical Services, Inc.

For complete contact information please visit www.westpharma.com.

Copyright © 2015 West Pharmaceutical Services, Inc.

www.smartdose.com I (800) 345-9800

The SmartDose® electronic wearable injector

combined with the HealthPrize adherence program

makes for a powerful combination. The SmartDose

injector helps your patients leave the treatment

center behind, making self-administration at home

simple and easy. And while at home, HealthPrize

helps your patients stay on track with their

therapeutic routine, through rewards-based patient

education and adherence tracking.

Empower your patients

Advancing Care, Improving Lives

#9387 1115

Page 3: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Cover: Nicholas Eveleigh/Getty ImagesIllustration: Dan Ward

November 2015

Features

COVER STORY

18 Exploring the Potential of Continuous Coating

Industy experts share insights on the advances

in tablet coating technologies and the potential of

continuous coating in solid-dosage manufacturing.

PARENTERAL MANUFACTURING

22 Better Days for Parenterals?

Although shortages, quality, and regulatory

challenges remain, improved technologies and new

investments suggest that the worst may be over.

FORMULATION

26 Demonstrating Therapeutic Equivalence

for Generic Topical Products

The determination of topical bioequivalence

requires a multi-faceted approach, tailored

specifically to the generic-drug formulation.

SUPPLY CHAIN

35 Piloting Track-and-Trace Implementation

Virtual pilot programmes examine scenarios that may

occur while implementing serialization requirements.

PharmTech.com

Columns and Regulars

5 Editor’s Comment

Complexities of Compliance

6 European Regulatory Watch

EU’s New Telematics Strategy

for the Regulation of Medicines

10 Drug Development

Risk-Sharing Agreements in the EU

and Considerations for Moving Forward

14 Outsourcing Review

CMOs Continue to Improve

Overall Biomanufacturing Performance

33 API Synthesis & Manufacturing

Advances in Heterocyclic Chemistry for API Synthesis

37 Product/Service Profiles

41 Troubleshooting

Using Dynamic Thermal Imaging to Correct Sealing Problems

42 Ad Index

Peer-Reviewed28 Identifying Causes Of Delamination

A long-term study was conducted to determine

the causes of delamination. The findings enabled

the development of an improved manufacturing

process for glass vials and a new type of vial.

Join PTE’s communityJoin the Pharmaceutical Technology Europe group on LinkedIn™*

and start discussing the issues that matter to you with your peers.

Go to PharmTech.com/linkedin

* The linkedIn logo is a registered trademark of LinkedIn Corporation and its affi liates in the United States and/or other countries

28 3522 18

Pharmaceutical Technology Europe is the authoritative

source of peer-reviewed research and expert analyses for

scientists, engineers, and managers engaged in process

development, manufacturing, formulation and drug

delivery, API synthesis, analytical technology and testing,

packaging, IT, outsourcing, and regulatory compliance

in the pharmaceutical and biotechnology industries.

Online ExclusivesDiversifying the Global Heparin Supply Chain:

Reintroduction of Bovine Heparin in the United States?

The global supply chain for bovine and porcine

heparin and regulatory considerations are examined.

www.PharmTech.com/standards-regulation

FDA Overhauls Inspection Operations

New programme emphasizes quality, risk,

and global collaboration.

www.PharmTech.com/regulatory-watch-0

Advancing Development & Manufacturing

PharmTech.com

Pharmaceutical Technology Europe NOVEMBER 2015 3

Page 4: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

PharmTech Europe

Editor

Adeline Siew, PhD

[email protected]

PharmTech Group

Editorial Director

Rita Peters

[email protected]

Senior Editor

Agnes Shanley

[email protected]

Managing Editor

Susan Haigney

[email protected]

Manufacturing Editor

Jennifer Markarian

[email protected]

Science Editor

Randi Hernandez

[email protected]

Contributing Editor

Cynthia A. Challener, PhD

Global Correspondent

Sean Milmo

(Europe, [email protected])

Art Director

Dan Ward

Publisher

Michael Tracey

[email protected]

Associate Publisher

Chris Lawson

Tel. +44 (0) 151 353 3520

[email protected]

Senior Sales Executive

Stephen Cleland

Tel. +44 (0) 151 353 3647

[email protected] Sales Operations Executive

Barbara Williams

[email protected]

Published byAdvanstar Communications UK LtdHinderton PointLloyd DriveCheshire OaksCheshire CH65 9HQ, United KingdomTel. +44 151 353 3500Fax +44 151 353 3601

UBM Life Sciences:Chief Executive OfficerJoe Loggia

Executive Vice-President, Life SciencesTom Ehardt

Executive Vice-PresidentGeorgiann DeCenzo

Executive Vice-PresidentChris DeMoulin

Executive Vice-President, Business SystemsRebecca Evangelou

Executive Vice-President, Human ResourcesJulie Molleston

Executive Vice-President, Strategy & Business DevelopmentMike Alic

Sr Vice-PresidentTracy Harris

Vice-President, General Manager Pharm/Science GroupDave Esola

Vice-President, LegalMichael Bernstein

Vice-President, Media OperationsFrancis Heid

Vice-President, Treasurer & ControllerAdele Hartwick

UBM Americas:Chief Executive OfficerSimon Foster

Chief Operating OfficerBrian Field

Chief Financial OfficerMargaret Kohler

UBM PLC:Chief Executive OfficerTim Cobbold

Group Operations DirectorAndrew Crow

Chief Financial OfficerRobert Gray

ChairmanDame Helen Alexander

Kevin Altria

Associate Director,

Pharmaceutical Development

GlaxoSmithKline R&D

Reinhard Baumfalk

Vice-President, R&D

Instrumentation & Control

Sartorius AG

Rafael Beerbohm

Head of Quality Systems

Boehringer Ingelheim GmbH

Gabriele Betz

Department of

Pharmaceutical Sciences

University of Basel, Switzerland

Phil Borman

Manager, GlaxoSmithKline

Rory Budihandojo

Director, Quality and EHS Audit

Boehringer-Ingelheim

Christopher Burgess

Managing Director

Burgess Analytical Consultancy

Ryan F. Donnelly

Professor

Queens University Belfast

Tim Freeman

Managing Director

Freeman Technology

Filipe Gaspar

Vice-President, R&D

Hovione

Sharon Grimster

Vice-President, Development

F Star

Anne Marie Healy

Professor in Pharmaceutics and

Pharmaceutical Technology

Trinity College Dublin, Ireland

Deirdre Hurley

Senior Director, Plant

Helsinn Birex

Pharmaceuticals Ltd.

Makarand Jawadekar

Independent Consultant

Henrik Johanning

CEO, Senior Consultant,

Genau & More A/S

Marina Levina

Product Owner-OSD, TTC-

Tablets Technology Cell, GMS

GlaxoSmithKline

Roberto Margarita

Platform Director

Corden Pharma

Luigi G. Martini

Chair of Pharmaceutical

Innovation

King’s College London

Thomas Menzel

Menzel Fluid Solutions AG

Jim Miller

President,PharmSource

Information Services

Colin Minchom

Senior Director

Pharmaceutical Sciences

Shire Pharmaceuticals

Clifford S. Mintz

President and Founder

BioInsights

Ian Pearson

Senior Design Team Leader,

TSL Projects

Tim Peterson

Transdermal Product

Development Leader, Drug

Delivery Systems Division, 3M

John Pritchard

Technical Director

Philips Respironics

Thomas Rades

Professor, Research Chair in

Formulation Desgin and Drug De-

livery, University of Copenhagen

Jean Paul Remon

Ghent University, Belgium

Rodolfo Romañach

Professor of Chemistry

University of Puerto Rico,

Puerto Rico

Beatriz San Martin

Senior Associate

Field Fisher Waterhouse LLP

Siegfried Schmitt

Principal Consultant

PAREXEL

Stane Srcic

Professor

University of Ljubljana, Slovenia

Griet Van Vaerenbergh

GEA Process Engineering

Benoît Verjans

CEO

Arlenda

Andreas Weiler

Global Technical Sales Director

SAFC

Tony Wright

Managing Director

Exelsius

EDITORIAL ADVISORY BOARD

Above is a partial list of the Pharmaceutical Technology brand editorial advisory

members. The full board, which includes advisory members of Pharmaceutical

Technology North America, can be found online at www.PharmTech.com/EAB.

Pharmaceutical Technology publishes contributed technical articles that undergo

a rigorous, double-blind peer-review process involving members of our distin-

guished Editorial Advisory Board. Manuscripts for editorial consideration should

be sent directly to Susan Haigney, managing editor, [email protected]% PostConsumer

Waste

4 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Editorial: All submissions will be handled with reasonable care, but the publisher assumes no responsibility for safety of

artwork, photographs, or manuscripts. Every precaution is taken to ensure accuracy, but the publisher cannot accept

responsibility for the accuracy of information supplied herein or for any opinion expressed.

Subscriptions: Pharmaceutical Technology Europe is free to qualified subscribers in Europe.To apply for a free subscription, or to change your name or address, go to PharmTech.com, click on Subscribe, & follow

the prompts.

To cancel your subscription or to order back issues, please email your request to [email protected], putting PTE

in the subject line.

Please quote your subscription number if you have it.

List Rental: Contact Sarah Darcy; Tel. +44 1244 629 326 Fax +44 1244 659 321

Reprints: Reprints of all articles in this issue and past issues are available (500 minimum).

Contact Brian Kolb at Wright’s Media, 2407 Timberloch Place, The Woodlands, TX 77380. Telephone: 877-652-5295

ext. 121. Email: [email protected].

Copyright 2015. Advanstar Communications (UK) Ltd. All rights reserved.

No part of this publication may be reproduced in any material form (including photocopying or storing it in any medium

by electronic means and whether or not transiently or incidentally to some other use of this publication) without

the written permission of the copyright owner except in accordance with the provisions of the Copyright, Designs &

Patents Act (UK) 1988 or under the terms of a licence issued by the Copyright Licensing Agency, 90 Tottenham Court

Road, London W1P 0LP, UK.

Applications for the copyright owner’s permission to reproduce any part

of this publication should be forwarded in writing to Permissions Dept,

Honeycomb West, Chester Business Park, Wrexham Road, Chester, CH4 9QH.

Warning: The doing of an unauthorized act in relation to a copyright work

may result in both a civil claim for damages and criminal prosecution.

Page 5: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

EDITOR’S COMMENT

Complexities of Compliance

By July 2016, pharmaceutical companies in Europe will have to comply with the Identification of Medicinal

Products (IDMP) regulations, which were developed by the International Organization for Standardization (ISO)

as a means to globally harmonize the way medicinal products are referenced. IDMP standards provide the basis for

the unique identification of medicinal products—the aim is to simplify the exchange of data between pharmaceutical

manufacturers

across Europe and

facilitate regulatory

activities from

drug development,

registration, and product lifecycle

management to pharmacovigilance

and risk management. The reporting

requirements mandated under IDMP will

involve leveraging product information

from R&D, manufacturing, supply chain,

and commercial divisions. Successful

implementation could potentially

improve process and operational

efficiencies within an organization.

Nonetheless, the industry faces a

number of challenges in the run up to

the July 2016 deadline. Pharmaceutical

companies are well aware that

compliance is compulsory but timelines

are tight given the delay in the

finalization of the European Medicines

Agency’s implementation guidelines.

There are also cost implications that

must be factored in, for example, to

purchase the necessary software and

ensure that current data management

is under control. As IDMP should not

be considered an IT-only project to

standardize data across the company, it

is important that personnel from various

departments understand their roles and

responsibilities during the preparation

stage as well as for maintenance of IDMP

moving forward.

The regulation of medicinal products

has become increasingly complex over

the years, and IDMP implementation is

no exception. Non-compliance is not an

option for pharma because it comes with

financial penalties and in extreme cases,

marketing authorization licenses could

be revoked.

Adeline Siew, PhD

Editor of Pharmaceutical

Technology Europe

[email protected]

www.chemistry.umicore.com

Improvement is always on our mind. And in your mouth.

Of course we know that toothpaste is more useful in your

mouth than on a mirror, but sometimes we get carried away

with the brilliance of our formulas. Our Rhodium or Ruthenium

catalyst is used in the production of menthol for toothpaste and

chewing gum. Speaking of which, you should see the formulas

we pop out with gum.

Employee: Don Zeng/Sales and Business Development Manager

Innovation made. Easy.

Page 6: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

6 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Glo

be

: Zo

on

ar

RF/G

ett

y Im

ag

es

The European Union has embarked on an ambitious

programme to streamline the collection and use of data on

new and licensed medicines and their substances, including

excipients, throughout the EU. The scheme aims to ensure

that the European Medicines Agency (EMA), the EU central

organization for authorizing medicines throughout Europe,

and the licensing authorities of the EU’s 28 member states

will use the same IT system, based on a single data standard.

The objectives behind the programme are to improve the

efficiency of regulatory processes, raise cost effectiveness,

achieve interoperability for the sharing of knowledge, and

enhance collaboration through promoting open access to data

repositories.

The vision behind the scheme with details of its projects

and their benefits was outlined in a document (1) on an

EU telematics strategy and implementation roadmap for

2015–2017, published in August 2015 by the EMA and Heads of

Medicines Agencies (HMA) of the member states. The strategy,

however, is already running into problems, mainly stemming

from the complexity of the standards being applied to achieve

data uniformity and from variations in degrees of enthusiasm

among the members states for the scheme.

Most licensing agencies, called national competent

authorities (NCAs), are keenly supportive of the plan, but many

of them want to give precedence to pursuing their own IT

initiatives. A few are already refusing to participate in some

existing collaborative IT projects.

Industry views

In addition, the pharmaceutical industry is unhappy that it

has had minimal involvement in the creation of the strategy

while it is being offered, so far, limited participation in its

operation (1). The industry will not be engaged directly in the

EU telematics governance structure, which will be headed by a

management board representing EMA and HMA. Nonetheless,

the strategy document recognizes the “significant impact” of

the pharmaceutical industry, and is, therefore, allowing for its

participation in project working groups.

The main European pharmaceutical trade associations

are, nonetheless, expecting to be more of an active partner

in the achievement of  improvements to and the operation

of the telematics strategy. “There has been one meeting (so

far this year) between the EU telematics management board

and industry representatives,” said Remco Munnik, chair of

the telematics working group of the European Generic and

Biosimilar Medicines Association (EGA), in an interview with

Pharmaceutical Technology Europe.

“However, the question of the frequency and the scope of the

industry’s strategic contribution still remains open and a meeting

once a year may not be sufficient to achieve real progress on

this,” he continued. “An efficient telematics structure,  supporting

the EU regulatory network in its regulatory activities, is crucial

for the future. As an industry, we would like to be an active

partner, not only operationally but also strategically by being

involved in setting objectives that can be achieved.”

The industry is already participating in an EU task force

on the implementation of the International Organization for

Standardization’s (ISO) Identification of Medicinal Products

(IDMP), which is a key working group in realization of the

telematics strategy. The five standards of the ISO, which

comprise the IDMP, are at the core of the EU strategy. IDMP

provides the basis for the scheme’s aim of consistency,

uniformity, and single repositories where data on products and

substances can be reused throughout a medicine’s lifecycle. It

also has the potential to be the cornerstone of an internationally

harmonious data system, particularly since the US Food and

Drug Administration (FDA) is also planning to implement IDMP.

In the longer term, EMA expects that IDMP will provide the

EU with a pathway to the operation of a Global Ingredient

Archival System (GiNAS), a common substance identifier that

uses a consistent definition of substances throughout the

world. The National Centre of Advancing Translational Sciences

(NCATS), part of the US National Institutes of Health (NIH), is

currently developing the GiNAS.

The introduction of IDMP is proving to be a massive

challenge not only for EMA and the rest of the EU’s regulatory

network of NCA, but also for the pharmaceutical industry.

The EU is taking on a pioneering role because it will be the

first globally to implement the IDMP standards. The job is

enormous, especially with substances, mainly because of

the amount of existing data that has to be collated and, if

necessary, changed to comply with IDMP. The aim is that the

IDMP standardization of definitions of product and substance

concepts will substantially improve the identification and

exchange of information on medicines, not just within the EU

but also internationally.

IDMP implementation deadline

In the telematics strategy document (1), July 2016 is given as

the deadline for the start of the implementation of IDMP because

it is the date set in the EU pharmacovigilance legislation for

compliance with the standards. However, even when the

document was being published, the European Commission,

the EU executive responsible for the EU telematics strategy,

EU’s New Telematics Strategy

for the Regulation of MedicinesThe scheme aims to ensure that EMA and licensing authorities of

EU member states will use the same IT system, based on a single data standard.

Sean Milmo

is a freelance writer based in Essex,

UK, [email protected].

Page 7: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

ELIMINATE PARTICULATES

& FIBERS IN THE CORE.

www.sterile.com

SYNTHETIC WRITING SUBSTRATE

§ Low particulate

and non-shedding

§ Exceptionally durable

§ Tear, abrasion and

chemical resistant

§ Easy to write on

§ Double bagged

packaged sterile

HEPA FILTERED PRINTING SYSTEM

§ Print wirelessly into cleanrooms

§ Use with pre-sterilized

CLEANPRINT 10

§ 316L Stainless Steel

Construction, can be

completely disinfected

§ HEPA Filter cabinet

§ Sheet fed, high speed

digital printer using

chemical resistant ink

CUSTOM DOCUMENTATION

§ Logbooks, ID tags,

Forms and Labels

§ Constructed using

CLEANPRINT 10

§ Customized specifi cally

per customer

§ Individual unique

numbering

and integrity features

§ RFID Technology available

15 Lee BoulevardMalvern, PA 19355-1234 USA(610) 644-8335

Page 8: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

8 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

was being pressed to adjust the deadline so that regulators and

the industry would have more time to switch to IDMP.

Although there has been no official announcement,

the industry and software vendors understand that the

Commission has agreed that the IDMP will be introduced in

phases with the first step being the issuing in July 2016 of the

EU IDMP implementation guide. Also, the terms and controlled

vocabularies used with existing pharmacovigilance data will

by then be aligned with IDMP requirements. The submission of

pharmacovigilance data to IDMP standards will now start to be

mandatory in late 2017, according to the Commission.

“An important reason for the phased-wise approach is the

fact that the ISO standards are not yet final and will probably

only become final at the end of 2015 or early 2016,” says

Munnik. “[The] industry welcomes this approach. We should

rather take time and do this properly, instead of rushing and

resubmitting the data due to the short preparatory phase.”

“This IDMP data are available within industry, but might not

be in one place or structured,” he added. “In order to align

this data in a company, multiple departments and databases

will have to start sharing the same data. This is especially a

challenge for generic companies with large portfolios and

multiple manufacturing sites and partners.”

With substances, which include excipients as well as

active ingredients, the difficulty is that the data required by

IDMP, such as the characterization of chemical properties,

are not kept by the pharmaceutical manufacturers but

by external sources such as the contract manufacturers.

“There could be huge gaps in availability as well as quality

between the data held and controlled by the pharmaceutical

companies themselves and that held by external sources,”

Niels Henriksen, a consultant at the Danish IT service provider

NNIT A/S, told Pharmaceutical Technology Europe. “The data

required by IDMP, sometimes derived from work in the early

development phase of a drug, will be lying around somewhere

in a document. The difficulty will be finding it.”

The main vehicles for the introduction of the IDMP—at least

at the EU level—will be single data repositories, the biggest of

which will be that of the Periodic Safety Update Reports (PSUR)

set up under the EU’s current pharmacovigilance legislation. This

legislation requires marketing authorization holders (MAHs) to

provide updates on the safety profiles of the more than 500,000

medicines on the EU market. Another repository is EudraGMDP

on Good Manufacturing and Distribution Practice, giving details

on all GMP certificates and statements of GMP non-compliance.

Data integration and uniformity

An important stage in the implementation of the EU telematics

strategy will be when all application dossiers will have to be

IDMP-compliant and submitted in an electronic Common

Technical Document (eCTD). This requirement has been sched-

uled to be mandatory by 2018 for all application dossiers within

the EU’s centralized and decentralized licensing procedures.

This deadline, however, may have to be rescheduled following

the postponement of the start of the IDMP implementation.

A lot of the barriers facing the realization of the EU telematics

strategy at the member state level are derived from differ-

ences in priorities between EMA and the NCAs. EMA is required

mainly to implement EU legislation on medicines regulation

while the NCAs have a range of obligations including meeting

EU requirements and also carrying out national policies.

“In many cases,” the EU telematics strategy document (1)

acknowledges, “the NCAs have a variety of obligations outside

the pharmaceutical regulatory domain.” These obligations will

be dictated by public health needs, legislative mandates, and

economic conditions. As a result, a lot of NCAs will be giving

greater importance to their own IT projects and objectives.

The Netherlands Medicines Evaluation Board (MEB), for

example, is considering the option of working with groups of

other NCAs on data projects. “We are currently reconsidering

our own IT strategy and infrastructure, and keeping an open

eye on EU developments,” says Stan van Belkum, MEB deputy

director. “Wherever and whenever, the MEB will work with

colleague member states of similar size and adapt EU solutions

if proven functionally advantageous.”

The agency also wants to concentrate on improving its own

IT services, particularly those aimed at helping the industry

and some of which provide different services to those at the

EU level. “Since 2007, the MEB has a fully operational workflow

and document management system dedicated to all MEB

business processes in both national and EU procedures,”

says van Belkum. “On top of that, we provide marketing

authorization holders online access to information about the

status of their application procedures. For the coming years,

the MEB’s IT strategy aims to extend this portal with additional

functionality and to make part of the  information in that portal

publicly available—for example, assessment reports, product

information and relevant patient information.”

Within the EU’s medicines regulatory network, full data

integration and uniformity may not be accomplished for many

years, but for the industry, the priority will be to advance the

introduction of IDMP as far as possible not just for the sake of

reducing regulatory aacosts but also for its business opportunities.

Reference

1. HMA and EMA, EU Telematics Strategy and Implementation Roadmap

2015–2017, EMA/532765/2015 (London, August 2015). PTE

IDMP provides the basis for the scheme’s aim of consistency, uniformity, and single repositories

where data on products and substances can be reused throughout a medicine’s lifecycle.

Page 9: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

HETERO USA, INC. s 1031 CENTENNIAL AVENUE, PISCATAWAY, NJ, 08854 s (646) 226-1634 or email: [email protected]

Reaching new heights in contract manufacturing partnerships

Why HETERO?· 20 API and OSD facilities, most FDA-approved

including two in New York

· Value pricing through large scale, debt-free capacity in Special Economic Zones

· Vertically integrated manufacturing and development support from product discovery through commercialization

· Broad portfolio of ARV, ONC, CV, CNS and GI drugs

· Over 300 US DMFs and 150 ANDAs filed

For All Your Contract Manufacturing Needs, Think Hetero First.

Hetero is transforming contract manufacturing with large capacities

already funded by our existing generic business. With state-of-the-art facilities and a manufacturing-first business strategy, Hetero offers cGMP quality, customer-focused reliability and value-driven solutions to enhance your ROI.

Think Hetero First.Largest Global Antiretroviral Producer

HIV Malaria HepC

Highest ReliabilityHighest Capability

Highest Value

Page 10: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

(Sp

otl

igh

t im

ag

e)

Ima

ge

So

urc

e/G

ett

y Im

ag

es

Michael J.

Kuchenreuther, PhD

is a research analyst for

Numerof & Associates.

Michael N.

Abrams, MA, is

managing partner at

Numerof & Associates,

www.nai-consulting.com.

10 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

DRUG DEVELOPMENT

Risk-Sharing Agreementsin the EU and Considerationsfor Moving ForwardThe authors look at key factors driving the risk-sharing agreements

that have been implemented to reduce drug expenditures across Europe.

Payers across the globe are becoming increasingly

assertive regarding products whose costs aren’t in

line with perceived performance—a trend that shows

no signs of slowing down. In fact, Express Scripts, one

on the largest pharmacy benefit managers (PBMs)

in the United States, recently announced a plan that

would link the price of some cancer drugs to their

real-world performance (1). Express Scripts’ action is

significant, considering that the US has been slower

than many European markets to implement new

pharmaceutical reimbursement schemes.

Managed entry or risk-sharing agreements (RSAs)

represent another mechanism that payers use to

mitigate the cost of new drugs based on real-world

performance. Financial and performance-based RSAs

(PBRSAs) have been widely employed throughout

Europe to reduce drug expenditures. While these

agreements may not be universally applicable or

without challenge, they are gaining momentum in

certain markets where they will likely play a role in

enhancing economic efficiency.

This article explores some of the driving forces

behind RSA implementation, provides an update

on the status of RSAs in the European Union,

and discusses some of the inherent challenges

manufacturers will need to overcome to achieve

success under these types of arrangements.

Linking drug reimbursement to valueIn most realms of commerce, if a product fails

to deliver what’s been promised, consumers are

financially covered through refunds and warranties.

Consumers of pharmaceutical products and other

stakeholders, however, have no such protections.

Rather, the pharmaceutical industry has had a

longstanding model of selling products and getting

paid regardless of product performance. Perhaps

that explains estimates that 90% of conventional

medicines are not efficacious in 50–70% of cases (2).

In therapeutic areas such as oncology, which is often

characterized by high-priced drugs, the efficacy rate

with standard treatment has been reported to be as

low as 25% across all cancer types (3, 4).

Against this backdrop, global spending on

medicines is expected to grow to nearly US$1.3

trillion by 2018 (5). Government and commercial

payers in most developed markets face aging patient

populations and shrinking budgets. Not surprisingly,

they are increasingly calling for manufacturers to

“put more skin in the game” in return for broader

coverage. Contractual arrangements directly linking

reimbursement to a product’s effectiveness or

budget impact clearly represent one mechanism

through which risk can be shared between payers

and manufacturers.

In efforts to contain spending, public and private

payers are also shifting risk to healthcare providers

through alternative payment models such as

bundled payments and capitation. As these changes

progress, manufacturers must realize that they face

a future in which they will increasingly be required to

assume greater risks related to the performance of

their products.

Risk-sharing agreements have been largely confined to Europe, where payers have been more restrictive in their coverage, particularly of cancer drugs, and have used value calculations based on comparators and quality adjusted life years.

Page 11: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Quality isn’t developed overnight. We instill precision engineering, superior materials, and more than 40 years of experience into every one of our replacement parts.

Contact us today to experience the best trained and most knowledgeable customer service team in the business.

Choose the best. Choose NATOLI.

Natoli Engineering Company, Inc. 28 Research Park Circle, St. Charles, MO 63304, USAp: +1 636.926.8900 f: 636.926.8910� �!� �%!����!��&� �%!����!�

BETTER

Page 12: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Drug Development

12 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

The benefits of RSAs for payers are

quite apparent—cost containment,

efficiency (i.e., improved value for

money), and/or improved outcomes

for the covered population. There

are, however, potential benefits

for drug manufacturers as well,

including earlier market access,

differentiation, and the opportunity

for a premium price. Taken together,

it’s not surprising that RSAs continue

to gain attention from payers and

manufacturers and are becoming

a more integral part of leadership

objectives for both parties.

Trends in the growth of RSAs in the EU For years now, national health

authorities in European markets

have “hedged” their reimbursement

decisions with pricing controls and

negotiated price cuts and RSAs

to control costs. To date, RSAs

have been largely confined to

Europe, where payers have been

more restrictive in their coverage,

particularly of cancer drugs, and

have used value calculations based

on comparators and quality adjusted

life years. For instance, in addition to

successfully negotiating the lowest

price in Europe for Gilead’s Sovaldi,

the French government secured a

volume-based discount as well as a

money-back guarantee if treatment

doesn’t work (6).

Financially based schemes (e.g.,

price-volume agreements, price-

capping, volume capping, and

rebates) have been more widely

employed throughout Europe

compared to PBRSAs because

they are easier to implement and

track (7). In contrast, the design

and implementation of PBRSAs has

historically been more complex

due to the need for patient

follow up, lack of reliable data

generation/registration (e.g., lack

of infrastructure to track drugs or

non-responders), and increased

administrative burden for payers

and manufacturers alike. These

were just some of the clinical and

operational complexities that largely

characterized the United Kingdom’s

multiple sclerosis scheme in the

early 2000s, which proved to be both

costly and rather ineffective.

Nevertheless, RSAs, including

performance-based arrangements,

continue to be explored and

implemented in select EU markets

(see Table I). A recent review of

RSAs established between 1993 and

2013 identified 148 arrangements,

the majority of which were initiated

within in the past six years (8). Italy,

the UK, the Netherlands, and Sweden

accounted for approximately 71% of

these arrangements. In contrast, the

prevalence of RSAs in other major EU

markets such as Germany, France,

and Spain is markedly lower (2–3%).

Of the 148 arrangements,

approximately 36% included a

component that linked product

reimbursement to clinical outcomes

observed in the real world. Not

surprisingly, oncology was the most

active area for RSAs, likely due to the

number of new cancer drugs, the

high disease-related costs, and the

Table I: Examples of performance-based risk-sharing agreements (PBRSAs) from Italy and the United Kingdom.

Country Drug Indication Company Year Details

Italy

Vidaza

Myelodysplastic syndromes/

chronic myelomonocytic

leukaemia/acute myeloid leukaemia

Celegene 2010

Manufacturer provides 11% rebate

for patients not responding to three

cycles of the treatment.

Votrient Advanced renal cell carcinoma GSK 2011

Manufacturer pays for patients

not responding after 24 weeks of

treatment.

United Kingdom

Revlimid Multiple myeloma Celgene 2009

Manufacturer pays for patients

not responding after 26 cycles of

treatment.

Votrient Advanced renal cell carcinoma GSK 2011

Manufacturer provides an initial 12.5%

rebate with the possibility of future

rebates based upon the results of a

head-to-head trial against Sutent*.

Velcade Multiple myeloma Janssen 2007

Manufacturer refunds the full cost of the

drug for patients who experience less

than a partial response after four cycles.

*Note: The National Institute for Health and Clinical Excellence (NICE) revised this agreement in 2013, removing the head-to-head

trial component.

Source: Ernst & Young Biotechnology Industry Report 2013 and “List of Technologies with Approved Patient Access Schemes”

from the official site of NICE (9, 10).

Page 13: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Drug Development

[email protected]

www.anton-paar.com

Small, smart, powerful

MCP 150 polarimeters

The compact MCP 150 polarimeters

combine a small footprint with full

21 CFR Part 11 compliance,

including electronic signature, and

an accuracy of ± 0.005 °OR.

premium price tags typically associated

with these products.

A closer look at some of these

markets reveals certain nuances that are

important to call out. In the UK, there has

been a trend away from more complex

arrangements toward increasing adoption

of financial-based arrangements. In fact,

the majority of recent arrangements

were implemented as simple confidential

discounts (9). In Italy, RSAs have often

consisted of both financial-based and

performance-based components;

however, more recent arrangements have

included one or the other, not both.

In France, the aforementioned

Sovaldi example is just one sign that the

government is willing to expand the use

of performance-based arrangements. In

fact, Celgene committed to the French

government on the effectiveness of

their multiple myeloma drug Imnovid

in exchange for a higher price (11). The

agreement required Celgene to build a

registry for collecting real-world efficacy

and safety data.

Considerations for manufacturersPayers’ concerns with pharmaceuticals

in the current healthcare environment

include rising budget impact, differential

value, large patient populations with

difficulty in identifying responders,

and the uncertainty regarding duration

of treatment. At the same time,

manufacturers are concerned with

delayed market access due to prolonged

price and reimbursement negotiations,

demonstrating differentiated value in

crowded therapeutic areas, and earning a

premium price that is in-line with the value

their products create.

RSAs represent an appealing option for

both parties if designed appropriately and

implemented in a collaborative manner.

It’s true that these arrangements are

likely to increase administrative burden

for both payers and providers, particularly

PBRSAs that require patient-level data

tracking. It should also be noted that

PBRSAs bring inherent risks outside the

manufacturer’s direct control, including

inefficient healthcare delivery and poor

patient compliance. For these reasons,

RSAs do not represent a “one-size-fits-all”

solution.

Before entering into an RSA,

pharmaceutical companies should always

ensure that the potential benefits are

significant enough to justify the effort

and risks associated with these types of

agreements. In addition, manufacturers

must understand how their products

deliver value to ensure that the particular

measures agreed upon align with that

value story. Implementing RSAs requires

not only lengthy negotiations and

potentially costly administration, but also

the ability to efficiently capture real-world

evidence. As a result, pharmaceutical

companies must develop the appropriate

IT and data analytics capabilities to

successfully implement these types of

agreements.

References1. P. Loftus, “New Push Ties Cost of Drugs

to How Well They Work,” The Wall Street

Journal, 26 May 2015, www.wsj.com/

articles/new-push-ties-cost-of-drugs-to-

how-well-they-work-1432684755, accessed

28 Sept. 2015.

2. Cowen & Co: Therapeutic Categories

Outlook, 2008–2012 estimate, Cowen Group,

NY, USA (2008).

3. C. Womack, The Pathologist in Drug

Development, presentation at the Edinburgh

Pathology 2013 conference (Edinburgh,

June 2013).

4. I. Miller et al., Pers. Med. 8 (2) 137–148 (2011).

5. IMS Institute for Healthcare Informatics,

Global Outlook for Medicines Through 2018

(November 2014).

6. T. Staton, “France Strikes Big Hep C

Treatment Deal for Gilead’s Sovaldi,”

FiercePharma, 20 Nov. 2014, www.fiercep-

harma.com/story/france-strikes-big-hep-c-

treatment-deal-gileads-sovaldi/2014-11-20,

accessed 28 Sept. 2015.

7. J. Espín, J. Rovira, L. García, Experiences and

Impact of European Risk-Sharing Schemes

Focusing on Oncology Medicines. Granada,

Spain: European Medicines Information

Network (EMINET), Andalusian School of

Public Health, 2011.

8. J. Carlson et al., Appl Health Econ Health

Policy, 12 (3) 231–238 (2014).

9. Ernst & Young, Biotechnology Industry

Report 2013.

10. NICE, List of Technologies with Approved

Patient Access Schemes, retrieved

December 2014 from NICE, www.nice.

org.uk/about/what-we-do/patient-access-

schemes-liaison-unit/list-of-technologies-

with-approved-patient-access-schemes,

accessed 28 Sept. 2015.

11. C. Ducruet, “Médicaments: quand les la bora-

toires sont rémunérés à la performance,” Les

Echos, 3 March 2015. PTE

Page 14: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

OUTSOURCING REVIEW

(Sp

otl

igh

t im

ag

e)

Sto

ckb

yte

/Ge

ttyIm

ag

es

Biomanufacturing efficiency is on everyone’s

minds, being the single most important area

of focus for global bioprocessing. And contract

manufacturing organizations (CMOs) are on the

leading edge as they implement performance

improvements. CMOs must remain efficient if they are

to be competitive—so this is no surprise. Results from

BioPlan Associates’ 12th Annual Report and Survey

of Biopharmaceutical Manufacturing Capacity and

Production (1), offer some clues as to what CMOs are

doing to remain competitive.

CMOs’ love affair with single-use devices has been

well documented. Indeed, single-use implementation

and integration is a much larger focus for CMOs than

it is for biotherapeutic developers. And as the results

in Figure 1 indicate, it’s easy to see why: nine out of

10 CMOs agree that biomanufacturing improvements

over the past year are coming from the use of

disposable and single-use devices.

Given that CMOs have long been at the forefront of

single-use adoption, it’s perhaps more interesting to

look at factors that are rising in importance for CMOs.

One such factor is better process development, cited

by 81.8% of CMO respondents as contributing to

improved biomanufacturing performance, up from

two-thirds of respondents in 2014.

This is a notable result, as process development

outsourcing has been on the rise in recent years.

Separately, 43% of industry respondents reported

outsourcing at least some upstream process

development activities to some degree, up from just

17.1% back in 2010. Additionally, 41% reported at least

some outsourcing downstream process development

activities to some degree. Improvements in process

development, therefore, are an encouraging sign

for CMOs as this becomes a growing business

opportunity.

A similar pattern plays out in validation services.

This is also a growing area of opportunity for CMOs,

with validation services a more popular outsourcing

activity than process development. In the 2015

survey, for example, almost three-quarters (73%) of

industry respondents reported outsourcing at least

some validation services, up from less than two-thirds

in 2010.

Another area to which more CMOs attribute

internal performance improvements is upstream

production operations. In the 2015 survey, 64%

of respondents said that these improvements

contributed to better overall performance, up from

56% in 2014. In fact, CMOs were almost as likely

to credit upstream improvements as downstream

improvements with better biomanufacturing

performance. That may partly be due to the current

bottlenecks being experienced in purification and

separation operations. And CMOs’ experience with

multiple products and campaigns provide them

expertise that in-house manufacturers may not have.

Upstream biomanufacturing operations outsourcing

has been growing more rapidly than downstream

operations, according to BioPlan’s data. In the space

of five years, the percentage of industry respondents

outsourcing upstream operations has doubled, from

21% in 2010 to 42% in 2015. While outsourcing of

downstream operations has been on the rise, it hasn’t

had quite the same growth trajectory, up from 28% in

2010 to 39% of respondents in 2015.

Upstream operational improvements are less

of an industry focus for both CMOs and in-house

manufacturers. Indeed, when BioPlan surveyed

the industry on the single most important area or

operational focus in 2015, not a single CMO pointed

to upstream processing advances as the top area;

because downstream production (DSP) operations

issues remain strong. A worrisome 64% of CMOs said

that downstream processing is impacting capacity

and overall production by causing at least some

bottleneck problems (noted by 64%). In fact, only a

CMOs Continue to Improve Overall Biomanufacturing PerformanceBetter process development is creating industry benchmarks for bioprocessing.

OUTSOURCING REVIEW

Eric Langer is president

of BioPlan Associates,

tel. +1.301.921.5979, elanger

@bioplanassociates.com,

and a periodic contributor

to Outsourcing Review.

Upstream biomanufacturing operations outsourcing has been growing more rapidly than downstream operations.

14 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 15: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

www.rommelag.com

rommelag agP.O. Box · CH-5033 Buchs, SwitzerlandPhone: +41 62 834 55 55 Fax: +41 62 8345500E-mail: [email protected]

rommelag Kunststoff-MaschinenVertriebsgesellschaft mbHP.O. Box 1611 · D-71306 Waiblingen, GermanyPhone: +49 7151 95811-0 Fax: +49 7151 15526E-mail: [email protected]

rommelag USA, Inc.27905 Meadow Drive, Suite 9Evergreen CO 80439, USAPhone: +1.303. 674.8333Fax: +1.303.670.2666E-Mail: [email protected]

rommelag Trading (Shanghai) Co., Ltd.Room 104~107,1st Floor, No.510 Zhongshan South 2nd Road,Xuhui District, Shanghai, 200032 P.R. ChinaPhone: +86 21 5153 4967, +86 21 5153 4968Fax No.: +86 21 5153 4969E-mail: [email protected]

Advanced aseptic packaging in one operation cycleReliable – Simple – Cost-Effective

bottelpack® Technology:, Integrated clean room US-class 100, Recognized by GMP, FDA, JP …, Aseptic packaging of liquids, creams, ointments …, Endless container designs in PE, PP…

Your benefits:, Tamper-proof packaging, Easy to open, Simple to use, Shatter-proof, no splinter hazard

See us at COMPAMED Düsseldorf, November 16-19, Hall 8B, Stand N20

Page 16: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Outsourcing Review

Fig

ure

1 i

s c

ou

rte

sty

of

the

au

tho

r.

quarter of respondents are currently

enjoying no bottlenecks in their

downstream processing.

CMOs spending to offset potential capacity crunchNot surprisingly, CMOs are seeing

more problems than biotherapeutic

companies due to downstream

processing, and are experiencing

more significant production capacity

constraints too. The BioPlan study

indicated they will likely continue to

invest in better DSP technologies as a

way out of these problems, for example.

Facility constraints are the most

common factor CMOs cite as creating

capacity crunches at their facilities

over the next five years (cited by

more than two-thirds). Probably by no

coincidence, seven in 10 CMOs plan

to increase their spending on new

facility construction this year, by an

average amount of 11.3%. 

The next biggest culprit in projected

capacity constraints is downstream

purification capacity. Spending plans

for CMOs are positive: almost three-

quarters would be increasing their

capital equipment budgets, with an

average increase of 11.7%.

Expected budget hikes—for

capital equipment (11.7%) and new

facility construction (11.3%)—were

the largest of all areas tracked. To

grow their businesses, CMOs are

dedicating funds to offset potential

capacity constraints in the future.

Not surprisingly, better

downstream purification technologies

are also on the agenda. CMOs

note that downstream innovation

is the leading way to avoid future

capacity constraints. Spending

projections aren’t quite as buoyant

for downstream innovation, though

they are solid. In 2015, six in 10

will increase spending on new

technologies to improve efficiencies

and costs for downstream

production, for an average budget

increase of 6.1%. This is likely due

to new technologies providing more

incremental increases in efficiencies

as opposed to new equipment that

can quickly provide access to more

capacity and avoid crunches.

ConclusionSingle-use equipment is helping CMOs

achieve performance improvements,

both for downstream purification and

for manufacturing productivity overall.

But CMOs are taking numerous other

factors into account as they improve

efficiencies and lower costs. These

range from better analytical testing

and product release services to better

operations staff training, optimized

media and improved existing quality

management systems. Better process

development is also a growing area

of interest for CMOs as they take on

more process development work—both

upstream and downstream—for clients.

Nevertheless, one of the main

routes to overall productivity

improvements for CMOs will be

better downstream operations.

Besides the use of disposable

equipment, a majority of CMOs are

developing downstream processes

with fewer process steps. Many are

also using or evaluating a number of

technologies, including:

t� Membrane-based filtration

technologies

t� Ion-exchange membrane

technologies

t� Ion-exchange technologies with

higher capacity.

Biotherapeutic developers might

keep a close eye on these activities.

CMOs, with their broad experience,

multiple product lines, and need for

rapid changeovers, are often at the

forefront of innovation. Though their

requirements clearly differ from

those of biotherapeutic developers,

the process improvements sparked

by innovations and adopted by

CMOs can provide a recipe for the

industry as a whole. As such, it

will be interesting to monitor the

activities and technologies that

CMOs adopt to improve downstream

production operations and overall

biomanufacturing productivity.

Reference1. BioPlan Associates, 12th Annual Report

and Survey of Biopharmaceutical

Manufacturing Capacity and

Production (Rockville, MD, April 2015),

www.bioplanassociates.com/12th,

accessed 12 Oct. 2015. PTE

Use of disposable/single-use devices

Better process development

Overall better control of process

Improved downstream production operations

Better analytical testing & product release services

Improved upstream production operations

86.4%

83.3%

81.8%

66.7%

68.2%

77.8%

68.2%

72.2%

63.6%

66.7%

63.6%

55.6%

2015 2014

Source: 12th Annual Report and Survey of Biopharmaceutical Manufacturing, April 2015, www.bioplanassociates.com/12th

Figure 1: Improving biomanufacturing performance for CMOs, 2015 v. 2014 (Select Responses).

To grow their businesses, CMOs are dedicating funds to offset potential capacity constraints in the future.

16 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 17: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts
Page 18: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Nic

ho

las

Eve

leig

h/G

ett

y Im

ag

es;

Da

n W

ard

Tablets are coated for several reasons, such as to improve

appearance; for taste- and odour-masking purposes; to protect

the API from moisture, oxygen, or the gastric environment of the

stomach (e.g., using acid-resistant enteric coating); to separate

incompatible substances; and to control drug release, among others.

As the industry continues to explore the advantages of switching

from batch manufacturing to continuous processes, Pharmaceutical

Technology Europe spoke to Ali Rajabi-Siahboomi, chief scientific

officer at Colorcon; Charlie Cunningham, senior manager, Product

Development, Colorcon; Martin Hack, vice-president and general

manager, L.B. Bohle; and Hubertus Rehbaum, manager, Scientific

Operations, L.B. Bohle, to gain insight on the advances in coating

technologies and the potential of continuous coating in the production

of solid-dosage forms.

Tablet coating processPTE: What are the steps involved in a typical tablet coating

process? How do you control the different variables

affecting the quality attributes of the coated tablets, such

as uniformity and functionality of the coating?

Hack (L.B. Bohle): The coating processes currently used in the

pharmaceutical industry vary from simple taste- or colour-masking

up to highly complex multilayer functional coatings. But as tablets

become more sophisticated therapeutic systems (e.g., with controlled

release mechanisms or combined APIs), manufacturers must pay

Exploring the Potential of Continuous CoatingIndusty experts share insights on the advances in tablet coating technologies

and the potential of continuous coating in solid-dosage manufacturing.

A Q&A by

Adeline Siew, PhD

closer attention to the quality of

the coating. To achieve the required

high-quality standards, process

parameters such as air flow, spray

rate, spray gun position, and tablet

bed movement must be taken into

account. By using equipment with

reliable hardware components and

fine-tuned control loops, those

critical process parameters can be

kept in the design space, ensuring the

targeted product quality. In addition

to using laboratory analysis to

monitor the process, Raman or near

infrared (NIR) spectroscopy will also

play an important part in ensuring the

final product and process quality.

Rajabi-Siahboomi (Colorcon): In

my view, all tablets should be coated,

whether for trade-dress purposes

or for the functional properties

mentioned above. The tablet must

have a robust formulation with good

mechanical strength to withstand

the handling, transport, and film-

coating processes. Therefore, robust

formulation and physical design of

tablets are crucial for film-coating

success. Tablets with good hardness,

low friability, appropriate shape, and

low propensity for moisture uptake

are desirable.

The film-coating process is fairly

complex but can be broken down into

three primary control areas:

t� Coating application. The coating

dispersion is delivered to the

spray guns and atomized using

compressed air into fine droplets

for deposition to the tablet

surfaces. The viscosity of the

coating formulation, number of

spray guns (depending on scale of

coating pan), atomization, and air-

pattern conditions all play a role in

how smoothly and uniformly the

coating will be applied to the tablet

surface. The solids concentration

of the dispersion influences coating

process time, coating uniformity,

and ultimately, the quality of the

finished tablet surface.

t� Thermodynamics. The drying

air parameters of temperature,

humidity, and volume (air flow)

must be carefully balanced with

the spray rates for the liquid-

dispersion coating application.

As the coating dispersion is

applied continuously, maintaining

appropriate temperature and air

18 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 19: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Tablet Coating

volume versus the coating spray rate is important.

The right balance will prevent coating defects related

to over-wetting (tablet-to-tablet sticking), or over-dry

conditions (spray-drying).

t� Mechanical movement. Tablets are typically coated in

rotating, perforated drums, ranging in diameter from

10” at laboratory scale to more than 60” at production

scale, with batch sizes from a few hundred grams up to

several hundred kilograms. The speed of pan rotation

governs how many passes an individual tablet will pass

through the spray zone in a given coating cycle and has

a great influence on coating uniformity. Appropriate

design of baffles within the pan also facilitate tablet

movement and mixing to influence coating uniformity.

Scaling upPTE: What about scaling up the coating

process?

Hack (L.B. Bohle): When scaling up a

coating process, the three unit operations

of a film-coating process—which are mixing, spraying/

film building, and drying, operated simultaneously within

a coating pan during a film-coating process—must be

understood. Important parameters are the drum rotation

speed, the ratio of the air and liquid flows (as process air

flow and coating solution spray rate), and the tablet bed

temperature.

When scaling up, normally, the tablet bed temperature

or exhaust air temperature is kept constant. The reason is

to have the same film building temperature of the polymer

of the coating solution. The spray droplets spread on

the tablet surface, and film building is generated with a

certain temperature. Also, the ratio between mass flow

of process air and mass flow of coating solution (spray

rate) should be equal to have the same thermodynamic

condition for the drying. The drum rotation speed should

be scaled in a way that the tablet velocity—while tablets

move through the spray zone—is almost the same in

laboratory scale, pilot scale, and production scale. Other

parameters such as atomizing air flow and pattern air

flow and the distance of the spray nozzle tips to the tablet

bed surface need to be adjusted properly for even spray

performance in each scale.

Geometrical similarity of the selected coating drum

from laboratory to production size is useful too. Special

spray nozzles (laboratory, pilot, or production coater)

designed for different coater sizes (spray rates and droplet

sizes) are recommended for successful scale up.

Coating technologiesPTE: Can you tell us more about the recent

advances in coating technologies and their

impact on continuous coating?

Rajabi-Siahboomi (Colorcon): In the

past, continuous coaters were most often employed

for high-volume products with tablet throughput rates

in the range of 500 kg to over 1000 kg per hour. The

scale of equipment is now being modified to meet

lower production capacity demands, ranging from 50

kg to 500 kg per hour. New mechanisms have also been

developed to eliminate the start-up and shut-down

product losses associated with earlier continuous coater

equipment. Studies evaluating these improvements

conducted in an O’Hara Fastcoat continuous coating

system showed consistent colour uniformity for all

tablets from start-up through shut-down. Some machine

manufacturers have developed novel approaches to

coating in semi-continuous modes. In one example, the

GEA ConsiGma coater is able to coat small, 3-kg sub-

batches rapidly, with a high degree of accuracy. Using

this method, each sub-batch is able to be tracked from

the upstream continuous tableting process, and critical

coating attributes can be monitored in real-time. Use of

in-line, process analytical technology (PAT)-based Raman

analytics during coating ensures product quality of each

sub-batch.

Film-coating formulation technology is also advancing

with the use of lower-viscosity polymers that provide

significantly improved process efficiency compared with

more traditional hypromellose-based coating systems. A

benefit of low-viscosity coating formulations is the ability

for application of the coating dispersion at significantly

higher solids concentration compared to traditional

coatings, resulting in considerably shorter process times.

Rehbaum (L.B. Bohle): For the coating process,

two different approaches are currently being applied.

Some vendors have presented truly continuous coaters,

for which a continuous product flow into the long

coating drum and a continuous product flow out of the

Pharmaceutical Technology Europe NOVEMBER 2015 19

Page 20: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Tablet Coating

drum is characteristic. R&D at L.B.

Bohle, however, has shown that

at this point, a truly continuous

coating process does not meet

the requirements for functional

coatings. For tablet coating, the time

that each tablet is exposed to the

process directly affects the quality

of the coating. Therefore, a narrow

residence time distribution, ideally

a spike, is the key to achieving an

outstanding coating uniformity. As

the travel time of all tablet cores

through a truly continuous coater

cannot be controlled sufficiently

using inserts or other guiding

elements inside the drum, L.B. Bohle

has decided to follow the concept

of a fast-batch coating design with

our KOCO machine family. In this

approach, by optimizing the charging

and discharging as well as the coating

process in terms of spray rate, air

flow, and tablet-bed formation, we

are able to achieve a high throughput

at constant, spike-shaped residence

time distributions.

There is another reason to

decouple the coating process

from the upstream continuous

manufacturing of the tablet core,

which is the tablet core relaxation

time. For the majority of products, the

core needs to be given a sufficient

time to expand after the compression

process. Feeding the tablet cores

from the de-duster directly into a

truly continuous coater does not

allow the cores to expand before

applying the film coat. This implies

that the expansion of the tablet core

will continue with the film coat in

place, leading to cracks in the coating

layer. For functional coating, such

defects are unacceptable. As a result,

the tablet core expansion step, which

interrupts the continuous product

stream of a continuous production

line between the tablet press and

the coater, makes the advantage of

a truly continuous coater to keep the

product stream continuous obsolete.

Continous versus batch coating

PTE: How does continuous

coating compare with the

traditional batch coating?

Cunningham

(Colorcon): Compared to batch

coaters, the commercially available

continuous coating machines have

pan diameters that are half, or less,

than the diameter of manufacturing-

scale batch coaters; but the length

of the pan can be as long as 15 feet.

However, the resultant tablet-bed

depth, which is low, is more consistent

with laboratory or pilot-scale batch

coating pans; this shallower bed depth

ensures greater frequency of tablet

presentation to the spray zone.

In recent work, Colorcon studied

the effect of tablet residence time

and uniformity of tablet progression

and coating variability in a Thomas

Engineering Flex CTC continuous

coater. We were able to conclude

that individual tablets progressed

uniformly through the continuous

process at production rates of up

to 850 kg per hour with relatively

small variability in transit times and

superior coating weight and colour

uniformity compared to the traditional

batch coater process.

In another study conducted in a

Driam Driaconti-T continuous cycled

coater, we evaluated the performance

of a novel film-coating formulation

developed by Colorcon capable of

application at solids concentrations

up to 35% w/w solids. Although this

coater employs elongated rotating

drum technology, the drum is divided

into individual coating segments,

giving it the advantages of small-scale

batch production. In this coater, with

production rates of 110–180 kg/hour,

we also found that coating uniformity

was significantly improved over batch

coaters.

The common theme around all

currently available continuous and

semi-continuous coater technology is

a significantly reduced coating cycle

time and reduced exposure of the

tablets to the coating environment.

Rehbaum (L.B. Bohle): At this

point, we do not see advantages in

truly continuous coating, as indicated

previously. However, based on

experience with our KOCO machine

family, we can achieve shorter cycle

times and higher throughput, while

maintaining the coating uniformity.

PTE: What aspects of

continuous coating still

need to be addressed by

the industry?

Cunningham (Colorcon):

With continued innovation in

equipment design, some of the

past limitations in throughput rate

are being addressed. Equipment

is now available to meet a wide

range of production capacity

demands. Although current film-

coating formulation technology

provides satisfactory results in these

machines, new, low-viscosity coating

formulations are being developed to

maximize coating efficiency. Linking

continuous coating processes to

newly evolving continuous tablet

manufacturing lines is underway, and

continued technology development is

needed for ‘real-time’ PAT monitoring

of coated tablet quality attributes

to ensure seamless quality from

start to finish in continuous tablet

manufacture. Although there may

not be an issue with the regulators,

another aspect to be considered

is the definition of a batch while

manufacturing using continuous

processing. A batch can be defined

on a time or material lot basis,

depending on specific product

manufacturing flows. These may

be related to a mindset change for

manufacturers as well as the need for

clarification from the regulators.

Hack (L.B. Bohle): Currently,

truly continuous coating from our

perspective is limited to simple taste

masking or colour coatings, as long

as the tablet core is not required

to expand or the film coating has

highly elastic properties. One of the

limitations is because of the wide

residence time distribution, which

results in poor coating uniformity. To

be able to overcome this limitation,

new ways to control the flow of all

(100%) tablet cores are needed. Some

vendors have suggested solutions

with compartments and mechanical

barriers or inserts to guide the tablet

bed flow. Unfortunately, from our

experience, none of these measures

can ensure a spike-shaped residence

time distribution. The efforts to

overcome this limitation only provide

benefits if the coating process can

be an integral part of the continuous

material flow. If the expansion

behaviour of the tablet cores

forestalls the seamless transfer of

the core from tablet press/de-duster

to the coating process, there is no

benefit in using truly continuous

coaters. PTE

20 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 21: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Register for free at www.pharmtech.com/pt/thermal

EVENT OVERVIEW:

Proper drug release of high-dose hydrophilic drugs is a challenge

for formulators since both the initial drug burst release, as well as

the consequent sustained release, need to be controlled.

Excipients can provide an answer to drug release challenges when

their hydrophobic nature is coupled with thermal sintering. This

approach intensifies the retardation of drug release by forming

plastic matrices of high mechanical strength, which can withstand

dose dumping and achieve controlled release. In this webcast, a

formulation expert will:

■ Discuss BASF’s Kolliwax grades and their suitability for thermal

sintering

■ Describe how high-dose hydrophilic actives were selected to

demonstrate the efficacy of thermal sintering (60-65% w/w)

using Kolliwax grades.

■ Present processing parameters to enable customers to

reproduce controlled drug release of hydrophilic drugs using

thermal sintering.

For questions contact Sara Barschdorf at

[email protected]

Sponsored by

Presented by

ON-DEMAND WEBCAST Originally aired October 22, 2015

Thermal Sintering for Controlled Drug Release of Hydrophilic Drugs

PRESENTERS

Rajkiran Narkhede

Assistant Manager

Technical Services at BASF

Moderator

Rita Peters

Editorial Director

Pharmaceutical Technology

Key Learning Objectives

■ Learn how to control drug release of

hydrophilic drug using plastic matrices.

■ Review the sintering mechanism and

technology with provided examples.

■ Understand how to choose suitable

ingredients for sintering technology.

Who Should Attend

■ This webcast targets formulation

scientists, research scholars in the field

of pharmaceutical technology, polymer

industry professionals, as well as

graduate and postgraduate students in

pharmaceutical technology.

Page 22: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Ph

oto

dis

c/R

afe

Sw

an

/Ge

tty

Ima

ge

s; D

an

Wa

rd

The past five years have been extremely challenging for parenteral

pharmaceutical manufacturers. For commodity workhorse products,

the aftershocks of manufacturing and quality issues and pricing pressures

continue to be felt in operating rooms, neonatal facilities, ambulances,

hospitals, and healthcare facilities, particularly in the United States.

On the large-volume (i.e., 100 mL or higher volume) side, intravenous

fluids and compounds used for dialysis, nutritional supplementation, and

irrigation have been in short supply due to contamination, manufacturing

problems, or product discontinuation. On the small-volume side, supplies

of chemotherapy agents and anaesthetics may have emerged from the

crisis they faced in 2013, but continue to be tight.

In 2015, Baxter had to recall some basic large-volume parenterals

such as intravenous saline and dextrose, which were found to

contain particulates. The same problem had prompted the company

and other suppliers, including Hospira and B. Braun, to recall large-

volume parenterals in 2013 and 2014 (1). For large-volume products,

most of which are heat-sterilized, the major problem continues to be

particulates. Small-volume products are vulnerable to both microbial

and particulate contamination, which has been traced to glass and

packaging materials, and problems such as leaching and sorption.

As experts from the Parenteral Drug Association (PDA) have pointed

out (2), particulates are either extrinsic or intrinsic, based on whether

they originate from outside or within the project. Recalls called out

both types of particulate contamination. Between January 2013 and

June 2014, the Medicines and Healthcare Products Regulatory Agency

(MHRA) in the United Kingdom found that 11 out of 42 drugs listed on its

Drug Alert website were listed because of particulate contamination.

Between 2008 and 2012, particulate contamination issues led to 22% of

all injectable drug recalls (3).

Microbial contamination can have several sources. In an interview

with Pharmaceutical Technology Europe, consultant Barry Friedman

traces many recent cases to inadequate sampling plans and media fill

studies. Ideally, media fill studies should be designed to account for

worst-case conditions (4), and they should be repeated periodically

and adjusted for batch size.

Agnes Shanley

“With both small-and large-volume

parenterals, people often come up with

sampling plans based on small lots.

They do media fills and get acceptable

results, but then lot sizes increase,”

Friedman says, “and some of them

don’t increase the size of media fills to

a comparable degree.”

Another problem is failing to follow

United States Pharmacopeia (USP)

<71> guidance in the number of

samples taken as the drug moves from

Phases I to commercial production,

says Friedman. The number of

samples required is based on the size,

volume, and number of containers

to be filled (5). In addition, Friedman

notes the need to reduce the potential

impact of operators on the process

and product. Investing in isolators

can often be the best way to go, he

notes, as long as the equipment is

properly maintained and operators

are adequately trained to recognize

potential sources of problems. “It’s

important to check seals and to

examine sample ports and gloves for

cracks,” Friedman says.

Solutions to parenteral

manufacturing challenges focus

on technology, new materials, and

components. A number of companies

are using automated systems to

reduce the potential for operators

to contaminate product. Vetter AG,

a contract development and

manufacturing organization (CDMO)

in Germany, has invested EUR 300

million to more than double its sterile

fill and finish and other capacity (6).

The expanded sites are expected

to become fully operational in 2017.

Central to Vetter’s efforts is its

“Improved RABS Concept,” developed

inhouse, that combines the features of

an isolator with those of a restricted

access barrier system (RABS). The

company’s Improved RABS Concept

creates a two-barrier design with

a walk-in isolator to reduce risk.

Cleanroom staff work in a Class 10,000

area that can be entered only after

proper gowning and passage through

air locks. The RABS contains the

filling line in an ISO-5 area, providing

redundancy so that problems can be

detected, contained, and fixed before

they affect product (7).

Vetter’s equipment includes

an automated hydrogen peroxide

decontamination module, and can

22 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Better Days for Parenterals?Although shortages, quality, and regulatory challenges remain,

improved technologies and new investments suggest that the worst may be over.

Page 23: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Parenteral Manufacturing

Pharmaceutical Technology Europe spoke with Miriam Beyer,

European marketing manager, West Pharmaceutical Services,

Germany about the company’s parenteral business.

PTE: The past few years have seen manufacturing issues and

severe shortages of both small- and large-volume parenterals.

What are the reasons behind these problems?

Beyer: The US Food and Drug Administration has recognized

that drug shortages are a critical issue for the healthcare industry.

Approximately 37% of shortages are due to manufacturing issues.

Other major factors causing drug shortages are issues associated

with raw materials (27%) as well as delays and capacity difficulties

(27%). Drug shortages can result in problems ranging from the wrong

expiration date on the package to particulates in the drug product or

sterility issues with injectable drugs (see Table I).

PTE: What are some best practices for maintaining sterility and

avoiding non-compliance for each type of parenteral?

Beyer: Maintaining sterility is vital to avoiding the main causes of

drug shortages. Primary packaging plays a pivotal role in ensuring

sterility, since it is in direct contact with the drug and poses risk of

interaction if the packaging is not compatible with the drug product.

Manufacturing quality can be affected by several issues, including

bacterial endotoxin, particulate matter and bioburden. One example

of the impact is well reflected in an FDA guidance recommending

‘the time between washing, drying (where appropriate), and

sterilizing should be minimized because residual moisture on the

stoppers can support microbial growth and the generation of

endotoxins’ (1). This can cause a challenge if primary packaging

components are not available already sterilized.

Achieving lowest visible and subvisible particulate levels is

currently a hot topic. While associated with cleanliness, it is a key

factor today to meet product target profiles and ensure compliance.

Close collaboration between a pharmaceutical manufacturer

and its drug packaging partner is key to ensuring that high quality

standards are maintained throughout the manufacturing process.

In terms of packaging, it is important that both parties agree upon

the validation processes that will be used during manufacturing

in order to establish—and adhere to—quality specifications for

primary packaging components. Validation processes can have

significant benefits for drug manufacturing. For instance, a state-

of-the-art validated washing process must reduce endotoxin

content by at least 99.9% (a 3-log reduction).

Sterilization processes are generally also validated to meet

sterility assurance levels. Furthermore, a sterilization process

validation is maintained through appropriate change control and

revalidation/periodic review programmes, all of which comply with

applicable cGMP requirements.

There are, however, additional variables that play a role in meeting

best practices. Product configuration and formulation, for instance,

may have an impact on quality, as do bioburden resistance and

weight, along with bag material. It is also important to note that

the packaging of the actual components in question should also be

evaluated to determine whether a component/packaging format

meets the necessary regulatory specifications.

PTE: What role can packaging play in preventing these problems?

Beyer: Packaging plays a significant role in maintaining the

quality of a drug product and preventing impurities. To minimize

interaction with the drug, it is essential that the packaging

components’ design and formulation meet the containment

requirements mandated by the physical and chemical attributes of

the drug. Different drug products have different packaging needs,

and what works for one product might not necessarily be the best

choice for another. Selecting the right closure for a particular drug

product is crucial to ensuring that the drug reaches the patient

safely and with the intended therapeutic effect.

To ensure the perfect match between a drug and its packaging,

it is important for pharmaceutical manufacturers to work closely

with experienced packaging partners from the early stages of drug

development through production. Having a reliable packaging

partner allows pharmaceutical companies to focus on their core

competencies: bringing valuable drug products to the marketplace.

PTE: Can you please describe your company’s technology, its

history, how it was developed, and how it is being applied?

Beyer: West was founded in 1923. Its early efforts pioneered the

safe distribution of penicillin, insulin, and other life-saving drugs.

Today, approximately 110 million West components and devices are

used around the world every day.

West has a long-standing partnership with Japan’s Daikyo Seiko,

a manufacturer of high quality pharmaceutical delivery system

components. Daikyo has built a strong reputation for materials and

manufacturing innovation. Our partnership with Daikyo has enabled

us to develop products that help customers mitigate drug product

development risks and enhance patient safety. These products

include: FluroTec coated serum and lyophilization stoppers, syringe

plungers, and tip caps, Daikyo Crystal Zenith vials and syringe barrels,

and ready-to-sterilize (RSV) components.

We recently launched Daikyo ready-to-use/validated (RUV)

components. We believe these components meet the highest

sterilization and quality standards, helping pharmaceutical

manufacturers comply with rigorous aseptic processing

requirements while reducing operational and capital resources.

PTE: Can you please elaborate on any innovative science/

methods used and results achieved so far?

Beyer: Daikyo RSV and RUV components are manufactured

using clean, high-quality elastomer formulations, and then washed

and sterilized as needed to help reduce the manufacturing foot-

print, streamline processes, outsource risks around component

preparation, and eliminate bioburden. Both Daikyo RSV and RUV

components are prepared to a tight particulate specification and

fully visually-inspected. The new Daikyo RUV offering, which takes

RSV components and steam-sterilizes them in a validated process,

offers pharmaceutical manufacturers the opportunity to focus on

the fill-finish with a high-quality, ready-to-use component.

Reference1. FDA, Guidance for Industry, Sterile Drug Products Produced

by Aseptic Processing: Current Good Manufacturing

Practice (Rockville, MD, September 2004).

Addressing parenteral manufacturing challenges

Table I: Trends in injectables shortages.

Year 2011 2012 2013 2014

All Forms 251 117 44 44

Injectables 183 84 35 30

Source: FDA, Drug Shortages Infographic,

www.fda.gov/Drugs/DrugSafety/DrugShortages/ucm441579.htm

Pharmaceutical Technology Europe NOVEMBER 2015 23

Page 24: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Parenteral Manufacturing

reportedly complete a sterilization

cycle in three hours. After successful

pilots, the company plans to

implement this module in all its

cleanrooms within the next few years.

In September 2015, Italian

equipment manufacturer Fedegari

opened a Technology Innovation

Centre in Philadelphia that features

advanced equipment and aims

to facilitate collaboration on

development of new processes (8). It

also launched a new website, www.

Sterilize.it, where specialists can

share data and collaborate.

Other companies are developing

prefilled technologies as a way to

improve ease of use and protect

product safety. On 3 and 4 November,

PDA will focus exclusively on this

topic with a specialized event, The

Universe of Prefilled Syringes and

Injection Devices.

Driving innovations include high

heat-resistant and non-leaching

plastics, new processes such

as blow-fill-seal (BFS), and new

configurations for bottles, closures

and caps. The CDMO, Catalent, used

quality-by-design (QbD) principles to

develop its glass-free Advasept vial

design to improve BFS systems. The

process aims to reduce particulates,

the number of process steps, and

the need for operator intervention,

thereby, reducing contamination risk.

By using medical-grade polypropylene,

the risk of glass delamination and

breakage is eliminated (9).

West Pharmaceutical Services, Inc.

has developed closure and capping

technologies to reduce risk (see

sidebar). The company established

a centre of excellence in Ireland in

2014, devoted to improving new

generations of this technology.

Companies are also investing in

new capacity and buying CDMOs that

have special expertise in working

with parenterals (see Table I). Pfizer,

for instance, bought Hospira in

September 2015 (10), while a number

of Indian pharmaceutical companies

have acquired contract manufacturing

organizations (CMOs) in the US.

Piramal Enterprises’ Pharma Solutions

bought Kentucky-based Coldstream

Labs earlier in 2015 (11). Coldstream

has special expertise in advanced

aseptic manufacturing equipment and

highly potent API (HPAPI) handling

and processing.

The large-volume parenterals

market is fairly staid, with competitive

pressures and low profitability. In

2014, some manufacturers withdrew

long-established products for financial

reasons. Small parenterals, however,

are seeing healthy innovation and

growth, according to Vivek Sharma,

CEO of Piramal Enterprises’ Pharma

Solutions, who summarized market

developments in CPhI’s 2015 Annual

Industry Report (12).

Over the past 15 years, Sharma

says, 900 new injectable drugs have

been approved, and another 2400 are

currently in the pipeline. Innovation is

being seen in oncology, particularly in

the use of targeted delivery systems

using liposomes, PEGylation, and

depot injections. Prefilled syringes,

which reduce safety risks, will

experience major growth, Sharma

predicts, with biologics accounting for

half of the injectables R&D budgets

for the top 15 pharmaceutical and

biopharma companies.

Generic injectables, which were a

US$37-billion market in 2013, should

grow to US$70 billion in 2020, he

says, while the need to reduce risk

will drive more pharma companies to

outsource parenterals development

and manufacturing to CDMOs

and CMOs. Currently, injectables

outsourcing is a $6-billion business,

and Sharma expects it to show 10%

annual growth for the next five years.

References 1. FDA, FDA Updates on Saline

Drug shortage, www.fda.gov/

Drugs/DrugSafety/UCM382255.

htm, accessed 20 Oct. 2015.

2. PDA, Industry Perspective on the

Medical Risk of Visible Particulates

in Injectable Drug Products, www.

pda.org/docs/default-source/

website-document-library/

publications/industry-perspec-

tive-on-medical-risk-of-visible-

particles-in-injectable-products.

pdf?sfvrsn=2, accessed 20 Oct. 2015.

3. S. Tawde, J Pharmacovigilance online,

http://dx.doi.org/10.4172/2329-

6887.1000e128, 5 Jan. 2015.

4. M. Akers, “Parenteral Preparation,”

Chapter 26 in Remington Essentials

of Pharmaceuticals, Ed. Felton, L,

Pharmaceutical Press, 2013.

5. E. Kastango, Understanding USP

71 Sterlity Tests and Extending

Beyond Use Data, http://asp.

pharmacyonesource.com/images/

simplifi797/USP71ExtendingBUD.

pdf, accessed 20 Oct. 2015.

6. Vetter, “Vetter Embarks on a 300 Million

Euro Investment Strategy for Further

Development To its Manufacturing

Sites,” Press Release, 30 Sept. 2015.

7. Vetter, “Improved RABS concept

combines two proven aseptic

processes,” www.vetter-pharma.

com/en/services-solutions-en/

vetter-answers/improved-rabs-

concept-combines-proven-aseptic-

processes, accessed 22 Oct. 2015.

8. Fedegari, “Official Opening of Fedegari

$2.9M State-of-the-Art Technology

Centre,” Press Release, 15 Sept. 2015.

9. Catalent, Advanced Aseptic

Processing, www.catalent.com/

index.php/offerings/A-Z-Offerings/

advanced-aseptic-processing/

10. Pfizer, “Pfizer Completes Acquisition of

Hospira,” Press Release, 3 Sept. 2015.

11. “Piramal Invests $30.6 Million

in Coldstream Acquisition,”

PharmTech.com, www.pharmtech.

com/piramal-invests-306-million-

coldstream-acquisition.

12. “Injectables to Show Double-Digit

Growth Through 2020,” PharmTech.com,

www.pharmtech.com/injectables-show-

double-digit-growth-through-2020. PTE

Table I: Outsourcing injectables: M&A activity in CMOs (in US dollars).Buyer Acquisition Approximate sale value

Pfizer Hospira $17 billion

Pfizer Innopharma $360 million

Hikma Bedford Labs $225 million

Sun Pharma Pharmalucence N/A

Mylan Strides Arcolab $1.6 billion

Piramal Coldstream Labs $30.65

Source: CPhI 2015 Annual Industry Report, Part 3, and PharmTech.com,

www.cphi.com/documents/129623/1844059/CPhI+Annual+Report+2015+-

+Part+I/57d8295a-a73a-444e-b593-c6fccdf00dae.

24 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 25: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Presenters

George Svokos

Chief Operating Officer

AMRI

James Grabowski, PhD

Associate Director of

Mergers & Acquisitions

AMRI

Moderator:

Agnes Shanley

Editor

Pharmaceutical Technology

EVENT OVERVIEW:

Merger and acquisition (M&A) trends in the contract

services and biopharma space are set to continue in the

coming years. Although M&A at contract manufacturing

organizations (CMOs) can be a stressful time for clients,

customers should know that M&As offer various

opportunities for continuous improvement and the

service providers that go through M&A integration most

frequently are likely to be among the best proponents of

the process.

Key Learning Objectives

■ Best practices in approaching integration planning

■ Find out why change management is vital during

acquisitions

■ Learn what you should be communicating during

integrations and when you should be talking to the

CMO

Sponsored by Presented by

Optimizing M&A Integration for Customer Success: The CMO Perspective

For questions contact Sara Barschdorf

at [email protected]

On-Demand WebcastOriginally aired October 29, 2015

Register for free at www.pharmtech.com/pt/maintegration

Who Should Attend:

■ Branded Pharma/Biotech

Companies

■ Generic Pharmaceutical

Companies

Page 26: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Ad

am

Ga

ult

/Ca

iaim

ag

e/G

ett

y Im

ag

es

For a new generic-drug product to be approved by a regulatory

agency, it is necessary to prove that it is bioequivalent

with a reference product. For orally administered dosage

forms, bioequivalence is demonstrated by determination of

the pharmacokinetic profile of the generic-drug product and

demonstration of similar drug absorption and drug elimination

characteristics compared with the reference product.

Demonstration of therapeutic bioequivalence for topical

dermatological products, however, is not as straightforward. Most

topical dermatological products are designed to deliver the drug to

the skin and not into the systemic circulation. Therefore, evaluation

of blood pharmacokinetics does not provide information on

drug delivery into the skin. A survey of international regulatory

bioequivalence recommendations for generic topical dermatological

products provides useful insights into currently accepted practices (1).

In summary, the approaches that can be used to demonstrate

bioequivalence are:

t� bioequivalence study with clinical endpoint

t� bioequivalence study with pharmacodynamics endpoint

t� in-vivo dermatopharmacokinetic study

t� bioequivalence study with in-vitro endpoint

t� waiver from bioequivalence study.

Demonstrating Therapeutic Equivalence for Generic Topical Products The determination of topical bioequivalence requires a multi-faceted

approach, tailored specifically to the generic-drug formulation.

Robert Harris, PhD,

is chief technical officer at

Juniper Pharma Services.

Almost all regulatory

authorities will require data from

a bioequivalence study with a

clinical endpoint (i.e., proving

that clinical performance of the

generic-drug product matches

that of the reference product) for

most topically applied generic-

drug products. These studies often

involve a large patient population (n

> 500) to provide sufficient data for

statistical evaluation. Cost and time

for conducting these studies can be

significant. Ironically, these studies

are often considered to be the least

accurate, least sensitive, and least

reproducible means of demonstrating

bioequivalence for topically-applied

products. Sometimes regulatory

authorities will also request blood

pharmacokinetics, if there are

safety concerns relating to possible

permeation of the drug into the

systemic circulation.

An alternative approach to clinical

endpoint studies, which is generally

accepted for topical corticosteroid

products, is the application of

the vasoconstrictor assay (VCA),

otherwise known as the human

skin blanching assay. This assay is

effectively a bioequivalence study

with a pharmacodynamic endpoint.

Corticosteroids absorbed into the

skin cause a whitening of the skin

due to a vasoconstrictive effect.

Measurement of skin whitening

(by use of a scoring scale or

chromameter readings) provides an

indication of the extent of absorption

and action of the corticosteroid.

Although there has been much

debate relating to the reliability of

data generated by this approach, it is

still widely accepted and less costly

compared with clinical endpoint

studies—however, it only applies to

topical corticosteroid products.

*O�WJWP�dermatopharmacokinetic

studies can be used to support

demonstration of bioequivalence

through measurement of absorption

of drug into the top layer of the

skin, the stratum corneum (SC), as

a function of time. Tape stripping

is used to determine the depth of

penetration of the drug at a given

time. This method of determining

drug penetration into the skin is

applicable to all therapeutic classes

of drugs. Nonetheless, acceptability

26 Pharmaceutical Technology Europe NOVEMBER�������PharmTech.com

Page 27: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

of this approach to bioequivalence

testing has been hindered because of the

variability in methods and study designs

between testing laboratories. Recent

advances to the dermatopharmacokinetic

principle have been the use of open flow

perfusion (OFP) or dermal microdialysis

(DMD). These methods involve positioning

an ultrathin, semipermeable vessel into the

dermis and then perfusing the vessel with a

buffer solution. A drug applied to the skin,

which permeates the SC, will diffuse into

the tiny artificial vessel, and continuous

measurement of drug transport into the

dermis can, therefore, be measured.

This approach, however, is not relevant

for products that are designed to deliver

drug into the SC only. To support JO�WJWP�

studies, it is usual to conduct additional

in-vitro studies to compare in-vitro

performance of the generic-drug product

versus the reference product (e.g.,

rheology, pH, droplet size) and also possibly

in-vitro release testing (IVRT) using Franz

diffusion cells to determine drug diffusion

through artificial membranes or into/

through skin.

In some cases, it may be appropriate

to apply for a waiver from conducting

bioequivalence studies—although

generally, this situation only applies to

topical solution products. To qualify

for a waiver, the drug substance and

the excipients in the generic-drug

product should be qualitatively (Q1)

and quantitatively (Q2) the same as in

the reference product. Often, it is also

necessary to demonstrate that functional

attributes (Q3) of the generic drug match

the reference product (e.g., pH, particle

size, viscosity, etc).

It is apparent that selecting the most

appropriate approach for demonstrating

bioequivalence for a topical generic-

drug product is not a straightforward

process. However, there are continuing

initiatives within the pharmaceutical

industry for harmonization in regulating

generic-drug products, particularly topical

dermatological products.

In 2012, the International Generic Drug

Regulators Pilot (IGDRP) project was

launched to support global harmonization

for the regulation of generic-drug products.

In 2013, a workshop was organized by

the Product Quality Research Institute

(PQRI) with the objective of addressing

the harmonization of regulatory practices

relating to topical drug products (2). One

of the conclusions from the workshop

was that determination of topical

bioequivalence requires a multi-faceted

approach, tailored specifically to the

generic drug/formulation. Subsequently,

a decision tree “strawman” was proposed

as a guide for deciding what is required

for determination of bioequivalence for a

given topical product. The “strawman” is

summarized as follows:

t� 1st option: Evaluate feasibility of

biowaiver based on formulation similarity.

t� 2nd option: Is the product amenable to

VCA? If so, evaluate feasibility of using

the VCA method.

t� 3rd option: Is the product amenable

to pharmacokinetic determination

(i.e., transdermal drug delivery)? If so,

conduct a pharmacokinetic study (alone

or with other supporting studies).

t� 4th option: If the site of action is

the SC/epidermis, evaluate clinical

endpoint/dermatopharmacokinetics/skin

permeability.

t� 5th option: If the site of action is the

epidermis/dermis, evaluate clinical

endpoint/dermal microdialysis/skin

permeability.

t� 6th option: If the site of action is below

the dermis, evaluate clinical endpoint/

dermal microdialysis/pharmacokinetics-

based bioequivalence study.

Furthermore, the European Medicines

Agency published a concept paper (3) on

the development of a guideline on quality

and equivalence of topical products,

which to a large extent, reflects the

recommendations from the PQRI (2013)

workshop.

In summary, the three bioequivalence

approaches that are currently consistently

accepted by regulatory authorities are

bioequivalence studies with clinical

endpoints, in-vivo pharmacodynamic

studies (in particular VCA for topical

corticosteroid products), and waivers

for topical solutions. Also, most require

pharmacokinetic studies if there are safety

concerns relating to systemic exposure.

However, it is refreshing to see that the

regulatory authorities are giving credence

to alternative science-based methods for

demonstration of bioequivalence, rather

than insistence on clinical endpoint studies.

References1. A.C. Braddy et al., ""14�+ 17 (1) 121-133

(2015).

2. A. Yacobi et al., 1IBSN�3FT �31 (4) 837-846

(2014).

3. EMA, $PODFQU�1BQFS�PO�UIF�%FWFMPQNFOU�

PG�B�(VJEFMJOF�PO�2VBMJUZ�BOE�&RVJWBMFODF�PG�

5PQJDBM�1SPEVDUT�(London, Dec. 2014). PTE

Page 28: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

28 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

PEER-REVIEWED

Identifying Causes Of Delamination Carol Rea Flynn, Dan McNerney, and Palak Shah

Even the exceptionally inert Type I borosilicate

glass is susceptible to attack by aggressive

chemicals, causing it to spall. This phenomenon in

primary packaging for parenteral products, which

is referred to as delamination, has increased since

the introduction of innovative biopharmaceuticals.

A long-term study was conducted to determine

the causes of delamination. Of the factors in

the manufacturing process that can affect the

propensity for vial delamination, this study identified

the forming temperature of the vial bottom as

the greatest correlation to the phenomenon. This

knowledge enabled the development of an improved

manufacturing process for glass vials and a new

type of vial.

Glass is recognized as the gold standard for primary

packaging of parenteral drugs. Glass is durable, inert,

clean, and transparent—properties that are important in the

manufacture of containers for sophisticated applications.

The composition of the glass used has been continuously

improved over time. With its high hydrolytic resistance,

borosilicate glass (Type I glass) has long replaced the simple

soda-lime glass for the packaging of parenteral products.

Nonetheless, even a material as resistant as Type I glass

can be tested to the limit by aggressive chemicals. When

packaging liquids have a particularly high or low pH value,

for example, an interaction with the glass can take place

in which the entire surface of the glass corrodes due to

the migration of alkali ions from the glass into the solution.

Another known phenomenon is when layers of glass

flake off in specific zones of the packaging product and

contaminate the solution. This process, which is called

delamination, is observed during the long-term storage

of high-pH solutions and solutions containing chelating

agents or buffer systems as well as during the storage of

biologics such as monoclonal antibodies. Figure 1 shows

an extreme case of delamination created in the laboratory,

not under realistic pharmaceutical storage conditions. A

series of product recalls in the United States has affected

all the major manufacturers of primary pharmaceutical

packaging made of glass. Delamination has become a

serious problem from both a medical and an economic

viewpoint. The detached glass flakes could potentially lead

to health problems in conjunction with intravenous or intra-

muscular injection, although no cases of this kind are known

yet. In addition, each product recall generates significant

losses for the pharmaceutical industry, especially in the

high-cost biologics sector.

Delamination testing

In view of the increasing number of product recalls, in early

2011, the US Food and Drug Administration instructed the

pharmaceutical industry, in collaboration with primary pack-

aging manufacturers, to come up with solutions for the dela-

mination problem. Joint meetings involving representatives of

the industry and the regulatory bodies were held to assess

the current state of knowledge and discuss the future course

of action. One year later, on this basis, the US Pharmacopeial

Convention (USP) presented a new draft of General Chapter

<1660> proposing test procedures going beyond those previ-

ously laid down (1). The use of these more aggressive test

Carol Rea Flynn is director of technical services, Dan McNerney is

director of engineering, and Palak Shah is engineering specifications

manager, all at Gerresheimer, Vineland, NJ. All correspondence should be

addressed to Marion Stolzenwald, [email protected].

Submitted: 31 March 2015; Accepted: 20 April 2015.

CITATION: When referring to this article, please cite it as C.R. Flynn,

D. McNerney, and P. Shah, “Identifying Causes Of Delamination,”

Pharmaceutical Technology 39 (11) 2015.

Ayd

An

Mu

tlu

/E+

/Ge

tty

Ima

ge

s

Page 29: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Pharmaceutical Technology Europe NOVEMBER 2015 29

Delamination

conditions is intended to facilitate more reliable predictions of

the tendency of primary packaging to delaminate.

In spite of these efforts, a host of questions remained

unanswered on the subject of delamination. Little was

known about the causes of the phenomenon at the time,

and there was no established industry standard for

methods of systematic investigation. A comprehen-

sive long-term study on the subject of delamination

was conducted in association with Alfred University

(New York, US). The first task was to develop a reliable

method for the qualitative and quantitative analysis of

delamination. The study then forced delamination under

laboratory conditions to identify the causes of this phe-

nomenon.

Methods

Detecting delamination. Five different solutions

(highly purified water; sodium chloride [NaCl] solution

with citrate buffer pH 7; and solutions with pH values

of 5.5, 8, and 9.5) were used in the study. The solu-

tions were tested for the presence of visible and subvis-

ible particles, extractables (i.e., chemical compounds

and inorganic elements), and pH changes. The iden-

tification of particle formation in the solution using

dynamic light scattering (DLS) proved to be inconclu-

sive. So, in addition to pH determination by μ-electrode,

the study focused in particular on the filtration of the

solution with subsequent energy-dispersive X-ray spec-

troscopy (EDX) of the filter. Extractables were ana-

lyzed using an inductively coupled plasma (ICP) and

micronebulizer. A hydrolytic resistance test in accord-

ance with European Pharmacopoeia (3.2.1A) and

US Pharmacopoeia (USP) <660> was also conducted

(2, 3). This test determines the resistance of the interior

surfaces to migration of ions out of a filled vial.

Data were also collected on the vials. Surface morphology

data collected by scanning electron microscope (SEM) was the

main method used over and above visual inspection. Stress in

the section of the vial particularly at risk to delamination was

also visualized using a polariscope. A methylene blue surface-

Figure 1: In delamination, detached

glass flakes contaminate the stored

medicinal product. The vial shows

an extreme case created in the

laboratory, not a typical case.

Glass forming time

and temperature

Glass forming

technology

Surface treatment

Type of

glass composition

Thermal sterilization

or autoclaving

Time and storage

conditions

pH drug product

Chemical composition drug/

drug formulation

(e.g. buffer)

Other factors

Annealing temperature

Figure 2: Vial production (left) and pharmaceutical processes (right) are both

potential triggers of delamination.

All

fig

ure

s a

re c

ou

rte

sy o

f th

e a

uth

or.

Page 30: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

30 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Delamination

staining test was conducted, but was

only of secondary significance.

Root-cause analysis. Once a reli-

able analytical procedure had been

developed as described in the previous

section, a design-of-experiment (DOE)

study was created to identify the root

cause(s) of delamination. To ensure

that all the potential influence varia-

bles were taken into account, a broad

approach was chosen encompassing

the relevant factors during vial produc-

tion and also during the subsequent

pharmaceutical processes, which are

illustrated in Figure 2. First, in the

area of material formulation, the two

borosilicate glass types (Type I class A

Gx33 and class B Gx51-V) were tested.

For control purposes, a batch of vials

made of molded glass was included

in the study. In each case, 3-mL and

10-mL vials were investigated. During

the production process, the effects of

different burner settings and the asso-

ciated forming temperatures were

determined. As both temperatures and

temperature gradients are high during

formation, these parameters were the

prime focus of the study. The effects

of three temperature steps during the

formation of the vial bottom were com-

pared, and the potential influence of

continuous and indexing forming proc-

esses was investigated. A further focus

of the study was the effect of treating

the vials with ammonium sulfate to

remove alkali ions from the surface

(pre-leaching) and, in this way, render

them more hydrolytically resistant.

On the processing side, the effects of

washing, sterilization, and depyrogena-

tion were also investigated.

Forty-eight batches with an approxi-

mate total of 100,000 vials were pro-

duced and delivered to Alfred University

for the study, where they were filled

with test solutions and stored at 5, 25,

and 40 °C. The degree of delamination

was investigated at the beginning of the

test and again after three months, six

months, and one year. Further examina-

tions will take place after three years.

On conclusion of the evaluations

made at the end of the one-year

storage period, an additional series of

tests using USP <1660> was conducted

with correspondingly higher solution

concentrations and temperatures as

follows (1):

t� 0.9% potassium chloride (KCl), pH 8.0,

2 h at 121 °C

t� 3% citric acid, pH 8.0, 24 h at 80 °C

t� 20 mM glycine, pH 10.0, 24 h at 50 °C.

For these additional vials, an eval-

uation was made after 2 or 24 hours

(as required by USP <1660>) and again

after 4–6 months.

Results and conclusions

Effect of time. After one year, detailed

results on the triggers and course of

delamination were seen. The destruc-

tion of the glass surface to the point

where particles become detached is

clearly a time-dependent phenomenon.

Delamination was more evident the

longer the glass came into contact with

aggressive solutions. Corroded surfaces

and detached glass flakes occurred

with increasing frequency during later

tests (four to six months and longer).

The changes observed during the

study under realistic storage conditions

were confirmed during the test series

in accordance with USP <1660>, which

involves more stringent test conditions.

Despite high solution concentrations and

extreme temperatures, no corrosion or

detached glass flakes were observed

during the test phase (2–24 hours), with

glass flakes only becoming detached

after several months, as shown in an

SEM in Figure 3, for example.

Effect of solution and glass type.

There were also differences in the spe-

cific effects of different solutions. In

conjunction with Gx51-V glass, KCl solu-

tion had the most pronounced dela-

mination effect, followed by solutions

with citrate buffer and glycine. With

Gx33 glass, however, the solution with

citrate buffer had the most pronounced

delamination effect, followed by solu-

tions with glycine and KCl.

Effect of treated glass. Treating the

vials with pre-leaching makes the sur-

face of the glass susceptible to dela-

mination. The most severe forms of

delamination were found in conjunction

with treated glass. Taking all sample

types together, filled with aggressive

solutions, treated vials showed statisti-

Figure 3: Delamination occurs during longer-term storage of aggressive

medicinal products: unimpaired glass surface (left), initial signs of pitting

(middle), then detachment of glass flakes (right).

Bottoming Burner

Stress Ring

Original Glass Tube

Prime ROI forVisible Flakes

Figure 4: When the vial bottom is formed, glass components evaporate and

condense at the stress ring; ROI is region of interest.

Page 31: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Delamination

cally significant higher occurrence

of delamination than untreated vials.

Treated glass should therefore be

avoided when packaging delamina-

tion-inducing medicinal products, no

matter how successful it has proven

to be in other areas of application.

Effect of forming temperature.

An interesting finding in this study is

that in the manufacturing process,

the factor that has the greatest cor-

relation to the propensity for delami-

nation is the bottom forming temper-

ature. It was observed that only the

heel area of the sample vials dela-

minated, and this area is subject to

particularly high thermal loads.

As shown in Figure 4, vial dela-

mination typically occurs in a clearly

defined region of interest (ROI)

between the heel stress ring and

the base of the vial due to the base

forming process. In this process, the

vial is turned upside down and the

upper part is heated to such a high

temperature that sodium, potas-

sium, and boron evaporate from the

glass matrix. The vapours released

condense on cooler parts of the vial.

At the same time, the temperature

conditions directly underneath the

base permit the formation of a sec-

ondary thin layer of glass with an

average thickness of 0.5 µm. This

process is abetted by the supply

of sodium and potassium in the

vapour, which lowers the melting

point. In contrast, the body of the

vial is so cool that no new layer of

glass can form. The surface layer of

glass at the stress ring is less inert

than the tubular glass and has a dif-

ferent thermal expansion coefficient,

which makes it more susceptible to

attack and degradation by stored

solutions. The central role of the

forming temperature in delamina-

tion also explains the initially sur-

prisingly result that the 10-mL vials

were more prone to delamination

than the 3-mL vials. Larger vials gen-

erally have greater wall thicknesses,

necessitating a correspondingly

greater supply of heat during for-

mation, which resulted in a greater

tendency to delaminate.

Cumulative % samples that delaminated

Bu

rner

tem

pera

ture

gu

ard

ban

din

gfo

r TH

OR

– 1

0 m

L V

ials

0 0,05 0,1 0,15 0,2 0,25 0,3

Figure 5: Correlation between burner temperature and delamination.

galenIQª Filler-BinderGreat choice. Great taste.

• Iso alt (Ph. Eur., USP-NF, BP, JPE)

• The filler-binder with sweet sugar-like taste.

• Your best choice excipient to be used for a broad variety of dosage for s.

Our team of experts is available for your product develop ent with galenIQ™.

Contact us for an appointment and a sa ple package.

[email protected] · www.galenIQ.com

ES691109_PTE1115_031.pgs 10.23.2015 02:54 ADV blackyellowmagentacyan

galenIQ™ Filler-BinderGreat choice. Great taste.

• Isomalt (Ph. Eur., USP-NF, BP, JPE)

• The filler-binder with sweet sugar-like taste.

• Your best choice excipient to be used for a broad variety of dosage forms.

Our team of experts is available for your product development with galenIQ™. Contact us for an appointment and a sample package.

[email protected] · www.galenIQ.com

Delamination

cally significant higher occurrence

of delamination than untreated vials.

Treated glass should therefore be

avoided when packaging delamina-

tion-inducing medicinal products, no

matter how successful it has proven

to be in other areas of application.

Effect of forming temperature.

An interesting finding in this study is

that in the manufacturing process,

the factor that has the greatest cor-

relation to the propensity for delami-

nation is the bottom forming temper-

ature. It was observed that only the

heel area of the sample vials dela-

minated, and this area is subject to

particularly high thermal loads.

As shown in Figure 4, vial dela-

mination typically occurs in a clearly

defined region of interest (ROI)

between the heel stress ring and

the base of the vial due to the base

forming process. In this process, the

vial is turned upside down and the

upper part is heated to such a high

temperature that sodium, potas-

sium, and boron evaporate from the

glass matrix. The vapours released

condense on cooler parts of the vial.

At the same time, the temperature

conditions directly underneath the

base permit the formation of a sec-

ondary thin layer of glass with an

average thickness of 0.5 µm. This

process is abetted by the supply

of sodium and potassium in the

vapour, which lowers the melting

point. In contrast, the body of the

vial is so cool that no new layer of

glass can form. The surface layer of

glass at the stress ring is less inert

than the tubular glass and has a dif-

ferent thermal expansion coefficient,

which makes it more susceptible to

attack and degradation by stored

solutions. The central role of the

forming temperature in delamina-

tion also explains the initially sur-

prisingly result that the 10-mL vials

were more prone to delamination

than the 3-mL vials. Larger vials gen-

erally have greater wall thicknesses,

necessitating a correspondingly

greater supply of heat during for-

mation, which resulted in a greater

tendency to delaminate.

Cumulative % samples that delaminated

Bu

rner

tem

pera

ture

gu

ard

ban

din

gfo

r TH

OR

– 1

0 m

L V

ials

0 0,05 0,1 0,15 0,2 0,25 0,3

Figure 5: Correlation between burner temperature and delamination.

galenIQª Filler-BinderGreat choice. Great taste.

• Iso alt (Ph. Eur., USP-NF, BP, JPE)

• The filler-binder with sweet sugar-like taste.

• Your best choice excipient to be used for a broad variety of dosage for s.

Our team of experts is available for your product develop ent with galenIQ™.

Contact us for an appointment and a sa ple package.

[email protected] · www.galenIQ.com

ES691109_PTE1115_031.pgs 10.23.2015 02:54 ADV blackyellowmagentacyan

Delamination

cally significant higher occurrence

of delamination than untreated vials.

Treated glass should therefore be

avoided when packaging delamina-

tion-inducing medicinal products, no

matter how successful it has proven

to be in other areas of application.

Effect of forming temperature.

An interesting finding in this study is

that in the manufacturing process,

the factor that has the greatest cor-

relation to the propensity for delami-

nation is the bottom forming temper-

ature. It was observed that only the

heel area of the sample vials dela-

minated, and this area is subject to

particularly high thermal loads.

As shown in Figure 4, vial dela-

mination typically occurs in a clearly

defined region of interest (ROI)

between the heel stress ring and

the base of the vial due to the base

forming process. In this process, the

vial is turned upside down and the

upper part is heated to such a high

temperature that sodium, potas-

sium, and boron evaporate from the

glass matrix. The vapours released

condense on cooler parts of the vial.

At the same time, the temperature

conditions directly underneath the

base permit the formation of a sec-

ondary thin layer of glass with an

average thickness of 0.5 µm. This

process is abetted by the supply

of sodium and potassium in the

vapour, which lowers the melting

point. In contrast, the body of the

vial is so cool that no new layer of

glass can form. The surface layer of

glass at the stress ring is less inert

than the tubular glass and has a dif-

ferent thermal expansion coefficient,

which makes it more susceptible to

attack and degradation by stored

solutions. The central role of the

forming temperature in delamina-

tion also explains the initially sur-

prisingly result that the 10-mL vials

were more prone to delamination

than the 3-mL vials. Larger vials gen-

erally have greater wall thicknesses,

necessitating a correspondingly

greater supply of heat during for-

mation, which resulted in a greater

tendency to delaminate.

Cumulative % samples that delaminated

Bu

rner

tem

pera

ture

gu

ard

ban

din

gfo

r TH

OR

– 1

0 m

L V

ials

0 0,05 0,1 0,15 0,2 0,25 0,3

Figure 5: Correlation between burner temperature and delamination.

galenIQª Filler-BinderGreat choice. Great taste.

• Iso alt (Ph. Eur., USP-NF, BP, JPE)

• The filler-binder with sweet sugar-like taste.

• Your best choice excipient to be used for a broad variety of dosage for s.

Our team of experts is available for your product develop ent with galenIQ™.

Contact us for an appointment and a sa ple package.

[email protected] · www.galenIQ.com

ES691109_PTE1115_031.pgs 10.23.2015 02:54 ADV blackyellowmagentacyan

Page 32: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

32 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Delamination

Improving vial production

This finding that vial delamination is

primarily triggered by the tempera-

ture conditions during vial formation,

in addition to the interaction of the

glass with the pharmaceutical product,

makes it possible to systematically

improve the production process. To

produce vials intended for the storage

of innovative or particularly aggressive

pharmaceutical drugs, formation below

a defined temperature limit takes pri-

ority over other factors, such as optical

perfection of the vial bottom. By imple-

menting quality-by-design principles in

the production operations, the input

parameters—in this case, the tempera-

ture of the bottom forming burners—

are continuously adjusted according to

the output on the basis of statistical

process control (SPC).

If the evaporation and recondensa-

tion of sodium, potassium, and boron

from the glass matrix is the most sig-

nificant cause of delamination, any

optimization of the production process

must start at precisely this point. The

most effective way to achieve this is

to optimize the forming temperature,

especially when smoothing the base

of the vial. During the study, vials were

produced across the entire forming

temperature spectrum. The cumula-

tive results across all other DOE vari-

ables show a clear correlation between

the burner temperature and the

number of delaminated vials, as shown

in Figure 5. Here, different process-

related temperature ranges were

taken into account for continuous and

indexing machines. However, the cor-

relation applies equally to both produc-

tion methods. It is, therefore, possible

to determine temperature limits for

each machine type below which dela-

mination in the ROI is greatly reduced.

This being said, there are still aggres-

sive solutions that are not recom-

mended to be used in Type 1 glass con-

tainers.

T h e T H O R p r o c e s s ( T h e r m a l

Hydrolytic Optimization and Reduction)

was developed by Gerresheimer to limit

the process temperatures and use an

improved bottom heat profile. A deci-

sive factor for the success of the opti-

mization is that THOR is designed as a

feedback SPC-controlled cycle.

In the process cycle, the forming

temperature (entire bottom matrix and

not a point sensor) of every single vial

is measured inline by camera systems

using proprietary software for the cam-

eras and subsequent data processing.

The results are forwarded to the

Infinity SPC system on the production

lines, which rejects all vials where the

forming temperature was exceeded.

In addition, the method of displaying

process statistics at the hot end was

modified so that an on-screen warning

signal is generated on occurrence of

excessive forming temperatures. The

forming parameters can be adjusted

in this case to keep the process within

the preset limits and reduce the reject

rates. Another positive side effect of

THOR cycle production is the improved

hydrolytic resistance of the vials. The

long-term study shows that vials pro-

duced within the defined formation

temperature limits fail the hydrolytic

resistance test far less often.

Quality-control test procedure

A p r o p r i e t a r y t e s t p r o c e d u r e

(Gerresheimer’s FLASH test) takes

into account the specific occurrence

of delamination in the stress ring near

the bottom of the vial. In this Ph. Eur.

3.2.1.A-compliant test, a vial is filled

with test liquid up to the nominal

volume and, after autoclaving, the

solution is titrated to determine the

hydrolytic resistance of the vial. If,

however, the tendency for delamina-

tion of the vial is the focus of the test,

it makes sense to fill the vial only to

a level just above the ROI in order to

obtain precise, delamination-specific

test results. Figure 6 shows a high cor-

relation between the test results with

a reduced filling volume and the rate

of delamination of vials in the frame-

work of the study for two types of glass

(Gx33 and Gx51). This method can be

used as a quality-control test.

Next steps

As there is no reliable way to replace

long-term testing, the study will be

continued for another year to deliver

further results on the interaction of

stored pharmaceuticals and vials pro-

duced under different conditions.

References

1. USP General Chapter <1660>, “Evaluation

of the Inner Surface Durability of Glass

Containers” (US Pharmacopeial Convention,

Rockville, MD, 2012).

2. Eur. Ph. 3.2.1A, “Glass containers for pharma-

ceutical use” (EDQM, Strasbourg, France, 2013).

3. USP General Chapter <660>, “Container–Glass”

(US Pharmacopeial Convention, Rockville, MD,

2013). PTE

Cumulative % samples that delaminated

1,20

1,00

0,80

0,60

0,40

0,20

0,00

0,00 0,10 0,20 0,30

Gx51

Linear (Gx33)

Linear (Gx51)FLA

SH

valu

e f

or 1

0 m

L v

ials

Figure 6: Correlation of Gerresheimer FLASH test with reduced filling volume

and delamination for two types of borosilicate glass (Gx33 and Gx51).

Page 33: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

API SYNTHESIS & MANUFACTURINGM

ich

ae

l B

an

ks/

Ge

ttyIm

ag

es

The structural complexity of newer small-molecule

drug candidates is increasing, with many

containing several different heterocyclic

substructures and numerous stereogenic centers.

Preparing single isomers of these compounds can be

quite challenging and requires the use of atom-

efficient, low-waste, regio- and stereoselective

reactions, according to Dave Green, vice-president of

R&D with ANGUS Chemical Company. “A considerable

amount of effort is being consumed to expand the

utility of classic synthetic methods, often focusing on

enantioselective catalysis or combining steps into

what are now being called multi-component reactions

(MCRs),” he observes.

In particular, multi-component coupling reactions

for the generation of heterocycles in fewer steps

with high regioselectivity and reduced processing

times, cost, and waste are attracting significant

interest. A new cross-dehydrogenative,

heteroaromatic carbon–hydrogen (C–H) silylation

reaction catalyzed by the simple base potassium

tert-butoxide (KOtBu) is also a potentially useful

reaction for API synthesis.

Cascade reactionsOlder methods for the synthesis of heterocycles

typically relied on the stepwise construction of linear

frameworks followed by final cyclizations. “Today,”

says Green, “reaction conditions and catalytic

methods are being developed such that three or even

four reactive components can be simultaneously

combined, resulting in a cascade that leads to a highly

functionalized product.” Nitroalkanes are often one of

the building blocks used for the synthesis of nitrogen-

containing heterocycles.

Nitroalkane MCRsIn one example published by Maiti et al. in 2010 (1),

fully substituted pyrroles were prepared with five

independently selected substituents (four-component

reaction) in a completely regioselective manner. “It is

also notable that all of the starting materials used in

the reaction are readily accessible monofunctional

reagents,” Green notes. For reactions using

nitromethane, isolated yields were often in the

70 to 80% range, and typically higher than those

using nitroethane (1). The 3-ketopyrroles obtained

from the reaction are highly versatile intermediates

and can be used for a variety of synthetic

applications, according to Green.

Dehaen and co-workers reported a three-

component coupling reaction between nitroalkanes,

alkyl azides, and aldehydes that affords tri-substituted

triazoles, again in a regioselective manner (2). This

reaction proceeds via 1,3-dipolar cycloaddition of the

azide with a nitroalkene formed in-situ. Tazobactam is

one example of a current API that contains a

1,2,3-triazole moiety.

Heterocycles as in-situ intermediatesANGUS utilized MCR technology to synthesize

3-(N-methyl)-1-phenyl-1-propanol via a heterocyclic

intermediate. The three-component reaction of

formaldehyde and styrene with N-methyl

hydroxylamine, which was readily prepared by

selectively reducing nitromethane, proceeded through

an isoxazolidine intermediate that was ring-opened to

give only the desired regioisomer in nearly

quantitative yield. This direct route to 3-(N-methyl)-1-

phenyl-1-propanol is an important development,

because this compound is the key intermediate in the

manufacture of fluoxetine, according to Green. “By

using the MCR technique, isolation of the methyl

nitrone intermediate was avoided, preventing the

issues associated with its propensity to oligomerize,”

he explains.

Handling energetic materials One of the challenges when preparing nitrogen-

containing heterocycles via multi-component

reactions that require the use of nitromethane or

compounds like N-methylhydroxylamine (MHA) is the

reactivity of these substances. Nitromethane,

Efficient syntheses are possible using multi-component and

cross-dehydrogenative, heteroaromatic C–H silylation reactions.

Advances in Heterocyclic Chemistry for API Synthesis

Cynthia A. Challener

is a contributing editor to

Pharmaceutical Technology

Europe.

Pharmaceutical Technology Europe NOVEMBER 2015 33

Page 34: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

API Synthesis & Manufacturing

azides, and to a lesser extent MHA

are energetic materials, and special

handling requirements must be

considered. “Minimization of the

quantities of in-process and/or

isolated materials is an excellent way

to also minimize the potential energy

release at any given point in the

process,” Green says. He points to

the recent development of

commercially available micro-flow

reactors as providing both

laboratory- and production-scale

options. “At the lab scale, a variety of

process conditions and

stoichiometries can be evaluated in

order to define the safe operating

limits for reagent concentrations/

temperature/pressure with

essentially no potential energy

release risk. In turn, scaling the best

operating conditions in an analogous

configuration greatly minimizes the

risk associated with handling the

larger volumes needed for

commercial production,” he

observes. 

Interest in heteroarylsilanesThe substitution of carbon for fluorine

is recognized as an attractive method

for preparing drug candidates with

improved lipophilicity and other

attractive properties. More recently,

compounds containing silicon-carbon

bonds have attracted interest,

because such derivatives are also

often more stable and have improved

solubilities and pharmacokinetic

properties.

To date, heteroaromatic

organosilanes have been prepared

using two rather limited methods:

stoichiometric reaction of a

heteroaromatic organometallic

intermediate generated via a Grignard

reaction with a silicon electrophile

and catalytic C–H silylation mediated

by a rhodium or iridium catalyst in the

presence of an excess of a hydrogen

acceptor. The functional group

tolerance of these methods is

minimal. In addition, they require the

use of large quantities of reactive

and/or expensive reagents. As a

consequence, there is a desire for

more general routes to organosilanes

that are not only more cost-effective

and practical on an industrial scale,

but also provide access to novel

compounds.

Fortuitous discoveryStudents in the Grubbs and Stoltz

groups at the California Institute of

Technology recently stumbled on an

interesting reaction catalyzed by

KOtBu. They were investigating the

conversion of biomass to chemicals

via the breakage of carbon-oxygen

bonds using an iron catalyst,

potassium tert-butoxide, and a

hydrosilane as a hydride equivalent

and observed that a control reaction

with just KOtBu and the hydrosilane

gave silylated heteroaromatics as

byproducts (3).

Using 1-methylindole as a model

substrate, the reaction conditions were

optimized for the selective production

of a single organosilane in high yield. A

catalytic amount (1-20 mol%) of KOtBu

was found to be most effective, and

cross dehydrogenative heteroaromatic

C–H silylation occurs without the need

for an acceptor, resulting in the

generation of hydrogen gas as the only

byproduct. Extensive studies were also

performed to confirm that the KOtBu

was indeed the catalyst (3).

The reaction is noteworthy not only

because it is novel, but also because of

its scope, scalability, and the fact that

it is catalyzed by an inexpensive,

commercially available substance based

on potassium, which is an abundant

alkali metal and not a rare and expensive

precious metal. Furthermore, the

reaction proceeds under mild conditions,

has no complicated byproducts, the

product is readily isolated, and in many

cases, no solvent is required.

With respect to the scope of the

reaction, indoles with a variety of

substituents on the nitrogen (N)

molecule and at various positions on

the arene ring, and many different

electron-neutral and electron-rich N-,

oxygen- and sulfur-containing

heterocyclic compounds are

tolerated, although carbonyl groups

generally are not unless protected as

acetals. In addition, bromide, iodide,

cyano, and nitro substituents inhibit

the reaction, while fluoride, chloride,

trifluoromethyl, epoxide, N-alkyl

aziridine, pyridine, and tertiary amine

and phosphine groups do not (3).

Simple arenes also undergo the

dehydrogenative heteroaromatic C–H

silylation reaction, but the reactivity

depends on the substituents. Anisole

affords the ortho-substituted silylated

product, while toluene and similar

compounds undergo directing-group-

free C(sp3)–H silylation to generate

silylated benzyl derivatives. The

electronic nature of any oxygen

substituents also determines the

regioselectivity (on the ring or the

substituent) of the silylation (3).

Importantly for pharmaceutical

applications, the reaction appears to

be readily scalable; the neat reaction

of N-methyl indole with 1.5

equivalents of triethylsilane at 45 °C

was performed on a 100-gram scale

followed by simple filtration and

distillation, affording the desired C2

silylated product in 76% yield with

high regioselectivity (> 20:1) (3).

In addition, the facile silylation of

APIs was achieved to demonstrate

the potential for application of the

reaction for late-stage modification of

pharmaceutically relevant

compounds. The antihistamine

thenalidine and the antiplatelet drug

ticlopidine were successfully

subjected to the regioselective C–H

silylation reaction (58–68% yield with

high chemo- and regioselectivity) (3).

Therefore, this method should be

useful for the rapid synthesis of

compound libraries. Different

heteroarylsilanes obtained using the

KOtBu chemistry were also converted

to more complex molecules via

silicon-directed Suzuki–Miyaura

cross-coupling and Hiyama–Denmark

cross-coupling reactions to

demonstrate their synthetic utility (3).

References1. S. Maiti et. al., J. Org. Chem. 75 (5)

1674–1683 (2010).

2. W. Dehaen et al., Angew. Chem. Int. Ed.

53 (38) 10155–10159 (2014).

3. A. A. Toutov et al., Nature 518 (7537)

80–84 (2015). PTE

Multi-component coupling reactions for the generation of heterocycles in fewer steps with high regioselectivity and reduced processing times, cost, and waste are attracting significant interest.

34 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 35: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

He

nri

k Jo

nsso

n/E

+/G

ett

y Im

ag

es

The pharmaceutical industry faces complex issues in its effort

to meet the requirements of the United States Drug Supply

Chain Security Act (DSCSA) (1). Pilot programmes are needed to

determine the feasibility of solutions, but the number of pilots

required will be costly, in terms of money, time, and human

resources. Virtual pilots, which use computer software to project

or simulate physical pilots, can reduce this burden by providing

some measure of learning or proof that a particular set of solutions

would be scalable and workable.

Traditionally, databases, spreadsheets, dashboards, and test

systems are used to understand systems, processes, and information

and to predict the expected outcome of a physical pilot. Simulation

software, however, is a more efficient technique that can be used

to create and execute a virtual pilot for any process, including

implementing the DSCSA in the pharmaceutical supply chain.

Simulation software has traditionally been used in manufacturing

environments to replicate complex processes and provide a way

to compare variations on specific scenarios to gain insights beyond

algorithmic calculations found in spreadsheets. Due to the increase

in computing power, simulation software has become easier to

use and is increasingly seen in many industries, where it is used

to study processes within and between systems, departments,

companies, and industries, including flow of products, people, cash,

and information. Simulations can also address human behaviour, time

considerations, and the physical environment.

The software has matured to the point that it can be used during

the information collection phase of a process study; it can provide

meaningful results, both at a summary level and in the details and

Piloting Track-and-Trace ImplementationVirtual pilot programmes examine scenarios that may occur while implementing

serialization requirements of the United States Drug Supply Chain Security Act.

Robert Celeste

is a founding partner of

RC Partners healthcare

consultancy, rceleste@

rcpartners.biz, tel:

1.215.584.7374.

nuances of a process. Simulations

can be used to create multiple

scenarios to examine alternatives

before large amounts of resources

are expended to plan, develop, and

execute a physical pilot. In fact, the

virtual pilot can become the blueprint

for the physical pilot.

Considering space and time constraintsCertain trading partners have

physical space or time constraints

that must be considered while

implementing DSCSA requirements.

Production lines, for example, vary

in terms of space available to include

serialization equipment. Simulation

can provide insight into how best to

use existing space, such as whether

to serialize on the production line

or serialize label-stock prior to the

production run. Because case and

pallet packing may be accomplished

elsewhere in the facility, a simulation

of the environment could help

address product and staff flow.

Likewise, wholesalers may have

time constraints in their window of

operations for picking and packing

orders to be delivered the next day.

Timing and positioning of product

and staff can be simulated to give

projections of what to expect upon

scale-up.

In addition to product and staff

flow, information flow is crucial

to meeting DSCSA requirements.

Creating, supplying, and archiving

DSCSA data can be simulated in

order to gain experience with the

latency, scalability, and feasibility

of making data available from other

systems within the organization.

All of these flows can be studied

in one simulation, to gain better

insight on how they interact with

each other and depend on each

other. For example, the DSCSA law

sets requirements for the timing of

transaction information. A virtual

pilot may be created to examine the

process flow of information, product,

and staff, should the product arrives

prior to the required Transaction

In addition to product and staff flow, information flow is crucial to meeting DSCSA requirements.

Pharmaceutical Technology Europe NOVEMBER 2015 35

Page 36: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Supply Chain

Information, Transaction History, and

Transaction Statement.

Another potential problem is

that during the transition to 100%

serialized products, companies

in the supply chain may receive

some serialized product and some

non-serialized product. To better

understand the transition, simulation

techniques could examine varying

numbers of products received, the

mix or percentage of serialized vs.

non-serialized products on hand, and

daily variability of serialized product

and associated traceability data.

Simulations can also examine the

latency of delivery and number of

suppliers.

Assessing human factors For complex systems or processes,

it’s often difficult to assess the

impact of individual tasks or even

understand these tasks and their

variation throughout the workweek

with enough clarity to confidently

declare that a pilot and ensuing

implementation will be successful.

Operations under the various stages

of DSCSA will require supplier,

customer, and internal process

changes as new steps are added to

existing processes, such as receiving,

picking, packing, and shipping. A

virtual pilot could demonstrate how

these new processes will affect daily

tasks as well as identify what changes

to support systems must be made.

Simulations could also provide insight

as to the activity balance needed to

keep up with throughput of product

and information.

A study of process and information

will include interviewing or speaking

to the people that perform the

process. It is imperative that they are

actively engaged in the information

collection and analysis process. There

are often nuances of why particular

step occurs and opportunities to

explore with the process expert

how newly accessible information,

equipment, or adjacent processes

might improve their process. Some

of this information may be difficult to

analyze. Using a virtual pilot, however,

allows inclusion of behaviours,

preferences, and other information

points that can directly affect the

outcome of the study or pilot. Also,

the process expert often is drawn into

the simulation process because it is

interactive and easily understood. The

end result is a more robust picture of

the process under study that can be

viewed at many levels and replicated

with different scenarios (e.g., five

people performing a task instead

of four). Lastly, simulations can

take into consideration the physical

environment itself, such as the layout

of equipment and materials, size

of rooms, and distance between

process points.

Incorporating business changesBusinesses are incorporating DSCSA-

triggered changes into established

operations that are constantly

changing due to changes in business

practices and economic conditions

(e.g., new customers, the loss of

existing customers, acquisitions, and

divestitures). Virtual pilots based on

simulations are an economic way of

re-running DSCSA scenarios, given an

ever-changing business environment.

Adding a newly acquired warehouse

or system to the simulation, for

example, offers an opportunity to

take advantage of existing work and

test it against the new reality.

ConclusionUnlike a static diagram, a simulated

environment actually runs; processes

require certain input, and if that input

is not there, the process simulation

stops until that problem is fixed. Each

glitch that is fixed in the simulated

environment is a glitch that won’t

have to be fixed in the final physical

pilot, during implementation, or in

production.

Not every pilot requires an

enormous investment in time, staff, or

funds. A pilot may merely demonstrate

proof-of-concept or experiment with

connecting systems to those of trading

partners in a test environment. A

company may be interested in piloting

what to do with all the new traceability

data made possible due to the DSCSA

law. A simulation or virtual pilot is

capable of generating a lot of data and

regenerating it based on changes in

input parameters.

Implementing the DSCSA, with

its serialized traceability system

that is interoperable with thousands

of trading partners, is a complex

challenge. There are many issues yet

to be understood and many details

that need to be incorporated into a

company’s plans and investment.

There are also many benefits and

opportunities to be gained by early

adopters who realize the value that

a more connected and visible supply

chain brings. Virtual pilots, with

simulation at their core, allow for

better contingency planning, “what

if” analysis, and even training of staff

in developing an awareness of new

departmental dependencies and

contributions toward the new reality

that serialization brings.  

Reference1. R. Celeste, “Global Serialization: Could

Virtual Pilots Be in Our Future?” www.

pharmtech.com/global-serialization-

could-virtual-pilots-be-our-future,

accessed 12 Oct. 2015. PTE

Virtual pilots based on simulations are an economic way of re-running DSCSA scenarios, given an ever-changing business environment.

The Drug Supply Chain Security Act (DSCSA) became

law in the United States in November 2013, but the

requirements of the law are to be phased in over a

10-year period.

The first phase, implemented in 2015, included the

requirement for trading partners to share transaction

information. Individual item serialization takes effect

in 2017, and traceability is scheduled to be in place by

2023. Visit www.PharmTech.com for more articles on

serialization in the US and globally, such as:

t� Serialization: Are you prepared?

Why this time is different

t� Serialization Shake Out

t� Considering Full Serialization

More on serialization

36 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Page 37: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

PRODUCT/SERVICE PROFILESPRODUCT/SERVICE PROFILES

Anton Paar’s Pharma

Qualification Package

To identify, analyze, control, characterize or

check your raw materials, intermediates,

APIs and final formulations, benefit from

Anton Paar’s portfolio of instruments

covering polarimeters, refractometers,

density meters, viscometers, rheometers,

synthesis instruments, sample preparation

instruments, X-ray analysis systems and

zeta potential instruments.

Anton Paar understands your need for

completely traceable results and supports you

to meet the strict pharmaceutical regulations.

Make life easier in your lab with

t� traceability via a complete user

management system

t� audit trail functionality

t� GMP-compliant printouts and reports

t� electronic signature

t� fulfillment of the requirements of 21

CFR Part 11

t� Pharma Qualification Package for defined

instruments which complies with

pharmaceutical regulations (GMP, GAMP

5, 21 CFR Part 11, USP <1058> including

risk analysis and traceability matrix).

Anton Paar develops and provides

laboratory instruments and process

measuring technology for industry and

research that fulfills the requirements of the

pharmaceutical industry.

Anton Paar GmbH

www.anton-paar.com

[email protected]

Fast Dissolve Oral Dosing

of Macromolecules

Catalent’s Zydis® ODT fast-dissolve

formulation is a unique, freeze-dried oral

dosage form that disperses instantly in the

mouth, requiring no water. This may

enhance the molecule’s performance and

could contribute towards better patient

compliance.

With more than 20 products launched

in 50 countries, Zydis is the world’s

best in class, orally disintegrating tablet

(ODT) technology. Whether to advance

pharmacokinetics through pre-gastric

absorption, potentially improve patient

compliance, or add marketing advantage

to a valued brand, Catalent’s customers

draw on Zydis fast dissolve technology

to potentially enhance investment value

and accelerate a product’s potential.

Catalent has now expanded the

Zydis technology platform to overcome

traditional oral delivery challenges of large

molecules such as proteins, peptides,

viral vaccines and allergans. Zydis Bio

has the potential to deliver fast-dissolve

formulations of such therapies in a

patient preferred oral fast dissolve dose

form, in as little as three seconds.

Catalent Pharma Solutions

www.catalent.com

[email protected]

galenIQ™—The filler-binder

excipient with exceptional

taste properties

BENEO’s excipient galenIQ™ (Isomalt Ph Eur,

BP, USP–NF, JPE and approved in China with

an Import Drug License) is a range of water-

soluble filler-binders. Derived from beet

sugar it has a sweet taste and promotes a

pleasant, well-balanced gustatory profile

in pharmaceutical formulations. Due to

these unique sensorial properties, it is

an optimal choice for the formulation

of solid and liquid oral applications, and

especially those in combination with

active ingredients or plant extracts which

have a bitter and/or unpleasant flavour.

The galenIQ™ range includes specific

grades for direct compression, for wet

granulation and also for specialized

processing technologies such as hot

melt extrusion, roller compaction, pan

coating, and as a sugar-free starter core for

multiple unit systems. The grades for direct

compression are free-flowing and highly

compressible. Furthermore the unique

surface morphology of these grades prevent

segregation within a powder mixture,

thereby ensuring high content uniformity,

in both low- and high-dosage formulations.

Moreover the physical and chemical stability

of galenIQ™ safeguards the constancy of the

corresponding finished formulations, making

it an ideal choice for the manufacture

of tablets, effervescent, sachets,

lozenges, and syrups that taste good.

BENEO, part of the Südzucker

Group, is a member of the International

Pharmaceutical Excipients Council (IPEC)

and produces galenIQ™ under GMP

conditions for pharmaceutical excipients.

BENEO GmbH

www.galenIQ.com

[email protected]

Pharmaceutical Technology Europe NOVEMBER 2015 37

Page 38: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

PRODUCT/SERVICE PROFILES

MicroSphere Refiner™

from the PSL Sterile

Production range

PSL have developed the MicroSphere

Refiner™ for microsphere formulation,

designed for process scale-up technologies

from lab scale to commercial production.

Polymeric microspheres obtained from

various micro encapsulation processes

require unique handling that differs from a

typical filtration and drying operation. With

side and bottom filtration, classification,

steam-in-place, drainability, tiltable

sterile discharge, vacuum drying and a

high yield, the MicroSphere Refiner™

technology has been designed to meet

such criteria for aseptic processes.

PSL is proud to participate in this great

technological advancement that will

improve the wellbeing of millions of

suffering patients.

PSL is an award winning international

manufacturer providing quality innovative

technology with process development

expertise to the pharmaceutical,

biopharmaceutical, chemical and laboratory

industries since 1989.

PSL will showcase the GFD® (Labora-

tory Filter Dryer) and the tiltable Micro-

Sphere Refiner™ for small scale pro-

duction from the PSL Sterile Production

range at P-MEC India, Hall 6, stand F50.

PSL (Powder Systems Limited)

www.powdersystems.com

[email protected]

PRODUCT/SERVICE PROFILES

38 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Nexera UC unified

chromatography system

Shimadzu’s Nexera UC unified

chromatography system is the world’s first-

ever unified and fully automated instrument

combining supercritical fluid extraction (SFE)

with supercritical fluid chromatography

(SFC). The SFE/SFC/MS platform merges

quick and easy online sample preparation

with advanced chromatographic analysis

and high sensitivity detection.

The Nexera UC serves a wide range of

applications, e.g. food control, research

in biopharmaceuticals and environmental

analysis. It enables highly reproducible

extraction and stable analysis even of

unstable samples prone to oxidation or

dissociation if exposed to light or air.

Shimadzu Europa GmbH

www.shimadzu.eu

[email protected]

bottelpack® 430—compact

blow-fill-seal technology

What is expected from a compact and

flexible laboratory BFS machine? A minimal

mould size to keep the costs for trials and

stability tests, etc. with various mould tools

low and also the advantages provided by

the rotation machine technology (con-

tinuous parison, little plastic waste) of

the popular bottelpack® 460/461 models.

The result of this mix is the bottelpack®

430—a combined shuttle and rotation

machine. The basic principle of the rotation

machine technology with a continuous,

extruded, uncut parison forms the basis

for this machine. The mould follows the

continuously extruded parison during the

blow-fill-seal process. Once the mould

has been opened, it cycles back and is

positioned directly above the recently

produced ampoule bar to shape the next

block. This creates a continuous ampoule

strip that is fed into an external punch. The

bottelpack® 430 is ideal as a laboratory

machine and also for small batch sizes.

rommelag ag

www.rommelag.com

[email protected]

Page 39: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

PRODUCT/SERVICE PROFILESPRODUCT/SERVICE PROFILES

SmartDose® Electronic

Wearable Injector

Designed to deliver high volumes of

injectable drugs over an extended period of

time, the SmartDose® electronic wearable

injector is designed to make it easier for

patients to self-administer medication

and can help encourage compliance

with prescribed treatments. Thousands

of SmartDose injectors have been used

in clinical studies, where patients have

self-administered the established dose.

The single-use, electronic wearable

injector is adhered to the patient’s body,

usually on the abdomen, and is provided

pre-programmed to provide the required

dose. The SmartDose injector includes a

silicone-free Daikyo Crystal Zenith® drug

container that is commercially available in

the ready-to-use format. West developed

the SmartDose injector with extensive

human factors analysis to understand and

design for preferences and behaviors at

various stages of the patient journey

West Pharmaceutical Services, Inc.

www.westpharma.com

[email protected]

Pharmaceutical Technology Europe NOVEMBER 2015 39

Advanced Homogeneous

Catalysts

Umicore is a materials technology

group focused on applications where its

expertise can make the difference.

We understand the rapidly changing needs

of our clients worldwide and provide innovative

metal based chemistry, homogeneous

catalysts and APIs, commercial-scale

manufacturing and supply chain expertise

to enable current and future technologies.

We offer an integrated long-term

collaborative partnership approach to

research, development and commercial

manufacturing that makes innovation

possible and offers customers continued

support throughout the product’s life cycle.

Key to our success is understanding

customer-specific applications —

enabling us to translate ideas into

chemistry solutions that work for our

customers’ process technologies.

Our Advanced Homogeneous

Catalysts allow you to design and run

t� a stereoselective hydrogenation,

t� cross-coupling and amination reactions,

t� metathesis reactions

with high efficiency and selectivity.

Combined to Umicore’s precious

metal recycling technologies, they

enable maximum cost efficiency in your

processes. Contact us to see for yourself.

Umicore

www.chemistry.umicore.com

[email protected]

Cleanroom Documentation

Systems

VAI is proud to introduce a new line of

Cleanroom Documentation Systems.

We have addressed and solved the

challenges surrounding particulate and

fiber contamination in controlled areas

from GMP required documentation

by developing, CleanPrint 10, the

Core2Print, and Core2Write.

CleanPrint 10, is the synthetic, cellulose

free, low particulate, writing substrate.

It is extremely durable, yet pliable, while

being resistant to abrasion, chemicals,

and ink smearing. The Core2Print, is a

316L Stainless Steel HEPA filtered printing

system. The Core2Print prints wirelessly

onto VAI’s pre-sterilized CleanPrint 10

paper into the controlled area. Core2Write

is a line of custom documentation

featuring: logbooks, ID tags, forms,

and labels. All Core2Write products are

printed on CleanPrint 10 synthetic writing

substrate. Core2Write products available

are sterile, and quadruple bag packaged

in VAI ABCD Cleanroom Introduction

System. Each product is completely

customizable, based on customer specific

requirements, and offer traceability via

barcode or patented RFID technology.

Veltek Associates, Inc.

www.sterile.com

[email protected]

Page 40: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

PharmTech.com

NOVEMBER 2014 Volume 26 Number 11

MARKET REPORT

Germany Post AMNOG

TECHNICAL Q&A

Continuous Manufacturing

PEER-REVIEWED

Sublingual Formulations

QbD in

Parenterals

Addressing

Particulate

Contamination

Advancing Development & Manufacturing

PROMO CODE: PTEHA15

*Free to qualifying readers in Europe

Signature Date

Mr/Mrs/Ms First name Family name

Job Title

Company

Address

Postcode City Country

Tel.

Fax

Sign here if you wish to receive (continue to receive) Pharmaceutical Technology Europe FREE

Print your name and address

Please quote your reference number if you have it:

START OR RENEW YOUR FREE SUBSCRIPTION TODAY!z ot oG www.pharmtech.com — click on ‘Subscribe’ and scroll down to Pharm

Tech Europe, or

z

z

Fax back on (00)1-218-740-6417 or (00)1-218-740-6437

E-Mail

Please let us have your email address – so we can keep our renewal costs down

1. What is your company’s primary business? (Fill in ONE only)

100 ◯ Pharmaceutical Manufacturing

110 ◯ Biopharmaceutical Manufacturing

150 ◯ Ingredients (e.g. Raw Materials, APIs, excipients,

chemicals, water)

141 ◯ Contract Services

160 ◯ Drug Delivery/Medical Product and Device

Manufacturing (e.g. inhalers)

105 ◯ Engineering/Facilities/Construction

80 ◯ University/Academia/Education

90 ◯ Government (including regulatory agencies and bodies)

2. What is your function? (Fill in ONE only)

10 ◯ Research/Development/Formulation

20 ◯ QA/QC/Validation/Regulatory Affairs

33 ◯ Supply Chain

44 ◯ Manufacturing/Processing

45 ◯ Information Technology

60 ◯ Engineering

80 ◯ Corporate Management

90 ◯ Project/Purchasing/Procurement/Contract Management

95 ◯ Education Professional

3. Would you also be interested in Pharmaceutical

Technology Europe’s weekly e-newsletter?

(Your Email address is required below.) Y ◯ Yes

fo esahcrup eht esirohtua ro ,yficeps ,dnemmocer uoy oD .4

services, equipment, and supplies? Y ◯ Yes N ◯ No

?erutuf eht ni gniwollof eht fo yna esahcrup ot nalp uoy oD .5

(Fill in ALL that apply)

A ◯ Raw Materials

B ◯ Processing/Manufacturing Equipment

C ◯ Drug Delivery

D ◯ Packaging

E ◯ Cleanrooms & Contamination Control

F ◯ Laboratory & Analytical Equipment

G ◯ Process Automation & Control

H ◯ Outsourced Services

I ◯ Compliance & Validation/QA/QC

Advanstar Communications provides certain customer contact data (such as customers’ names, addresses,

phone numbers and e-mail addresses) to third parties who wish to promote relevant products, services and

other opportunities which may be of interest to you. If you do not want Advanstar Communications to make

your contact information available to third parties for marketing purposes, please contact us by emailing

[email protected] or by mail at Advanstar, Bridgegate Pavilion, Chester Business Park, Wrexham Road,

Chester CH4 9QH, United Kingdom.

Which version of Pharmaceutical Technology Europe would you like to receive?◯ Digital ◯ Print ◯ Both

Mail form to Pharmaceutical Technology Europe Advanstar Honeycomb West Wrexham Road, Chester CH4 9QH, United Kingdom, or

Photocopy this form — complete all sections— fax it back

Page 41: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

TROUBLESHOOTINGD

an

lea

p/G

ett

y Im

ag

es

TROUBLESHOOTING

Fabian Schapiro

is VP Marketing and Sales

of DIR Technologies,

www.dir-technologies.com.

Problems in an induction-sealing process can be identified and corrected in real time.

Primary packaging sealing integrity is crucial in

ensuring product viability and shelf life. The days

of sampling as the primary means of inspection and

analysis for the quality control of pharmaceutical

packaging may be nearing an end. One method

for non-destructive, 100% inspection of packages

is using dynamic thermal imaging to inspect the

integrity of induction-sealed bottles. Inspection

is performed through closed bottle caps without

damaging or degrading the product and without

reducing line speeds. Precise data can also be

collected to increase understanding of the process,

improve quality, and allow operators to detect and

amend issues in real-time.

This solution goes beyond the level of accept/reject

analysis to also indicate the source in the process that

may have caused the faulty seals, including problems

related to the raw material, the capper, or the sealer.

Incorrect settings can be quickly detected, allowing

the immediate removal of defective bottles from the

production line and adjustment of the source of the

problem. Problems can be corrected in real time,

before an entire batch is completed. This process

analytical technology (PAT) for the induction-sealing

process allows the input and output to be tracked,

enabling continuous process verification.

How dynamic thermal imaging worksAll objects have a certain temperature and emit

waves of energy, called infrared radiation (IR), invisible

to the naked eye. In general, hot objects emit more

energy than cold objects. A thermal imager creates a

viewable image from the IR waves, showing a “heat

picture” of a scene.

Thermal imaging was originally developed for the

defense industry and is regularly used for military

and security applications, as well as non-intrusive

inspection of plumbing, electrical systems, and

insulation. Combined with high-speed image analysis,

this technology provides unique monitoring and

detection capabilities that can be used to inspect

pharmaceutical primary packaging, specifically,

induction sealing.

Induction sealing process problemsIn the induction sealing process, as shown in

Figure 1, a bottle is filled, and a closure with

a composite liner inside is screwed onto the

filled container. The container then travels by

conveyor under an induction sealer, which uses an

electromagnetic field to heat the aluminum foil layer

of the liner. The heated foil, in turn, causes the heat-

seal layer to melt. When cooled, the polymer creates

a bond with the container, resulting in a hermetically

sealed product. When the cap is removed, the foil

remains, providing an airtight seal and preventing any

tampering or contamination of the bottle’s contents

until the customer pierces the foil. A good induction

seal prevents product leakage, is tamper-evident,

maintains product quality, and fosters consumer

confidence.

Maximizing the efficacy and efficiency of an

induction sealer requires a holistic, carefully

coordinated approach that incorporates machine,

materials, quality control, and operator education.

The container, closure seal liner, and induction sealer

must be compatible, and an operating window that

allows the sealer to be successful within a range

of predetermined operating tolerances must be

established. Defects can originate at various steps

along a packaging line, including the capper, induction

machine, conveyor and, of course, the raw material

itself. Potential problem areas are described as

follows:

t� Material-related problems include bent or

damaged foil; missing foil; damaged edge or edges;

or an upside-down liner (Figure 2a).

t� Problems related to the capping equipment include

untorqued or crooked caps (Figure 2b).

t� Problems tied directly to the sealer itself usually

involve overheating or under-heating (Figure 2c).

Underheating the foil can result in the familiar

Using Dynamic Thermal Imaging to Correct Sealing Problems

Pharmaceutical Technology Europe NOVEMBER 2015 41

Page 42: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

All

fig

ure

s a

re c

ou

rte

sy o

f th

e a

uth

or

Troubleshooting

‘potato-chip’ effect, as shown in Figure 2c. Overheating

the foil can cause damage to the seal layer and to any

protective barriers, which could result in faulty seals,

even weeks after the initial sealing process. Other

sealing problems do not typically originate with the

induction sealer but rather, are associated with a fault

earlier in the process that made it impossible for the

sealer to perform properly.

Torquer problem case studyIn a classic torquer problem, an incorrectly calibrated

torquer does not correctly close the cap, and as a

result, the induction sealer cannot create a proper seal.

Figure 3 shows thermal imaging results used to track the

process. In the first batch of 2000 bottles, a significant

number of rejections (shown as red dots) is seen. This

deviation from the defined process window indicates

that there is a faulty process that should be stopped and

the source of the problem checked before more bottles

are incorrectly sealed. The rejections in this case were

discovered to be associated with the tightness of the

cap. Based on this information, the line operator stopped

the line and adjusted the torquer. The batch was then

continued and rejections return to an acceptable number,

as shown in Figure 3.

ConclusionGiven the myriad potential problem areas, the need for a

troubleshooting process that assures the highest possible

level of quality control is clear. In each of these cases,

use of dynamic thermal imaging allows the operator to

identify the problem early, stop the line, immediately

remove any defective bottles, and make necessary

corrections to the production line process. Preventing

issues before they become significant problems minimizes

the cost of wasted time, work-backs, materials, and

labour. This real-time, non-intrusive monitoring of package

integrity is currently in use on packaging lines at several

leading pharmaceutical packaging companies. PTE

42 Pharmaceutical Technology Europe NOVEMBER 2015 PharmTech.com

Airbridge Cargo Airlines ..........................................................................29

AMRI .........................................................................................................25

Anton Paar ................................................................................................13

BASF ..........................................................................................................21

Beneo ........................................................................................................31

Catalent .....................................................................................................44

CPhI India ..................................................................................................17

Hetero Inc ....................................................................................................9

Natoli Engineering ....................................................................................11

Powder Systems .....................................................................................19

Rommelag .................................................................................................15

Shimadzu.............................................................................................27, 43

Umicore .......................................................................................................5

Veltek ...........................................................................................................7

West Pharmaceuticals ...............................................................................2

Ad IndexCOMPANY PAGE COMPANY PAGE

Figure 1: Steps of the induction sealing process.

Figure 2: Problems can be related to materials (a), capping equipment (b), and sealing equipment (c).

Bent / damaged foil Missing foil Damaged edge Upside-down

liner

(a)

Under-heating Overheating

(c)

(b)

Untorqued cap Crooked cap

Figure 3: Thermal imaging tracks the induction-sealing process and inicates rejected bottles (red dots).

Batch continued with an acceptable number

of untorqued caps

Number of Bottles

Torquer

related

parameter

Torquer

problem

solved

Torquer

problem

identified

EMPTYBOTTLE

FILLERCAPS

TORQUER

INDUCTION SEALER

Page 43: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

CHANGES EVERYTHINGShimadzu’s new LCMS-8060 makes a real differ-ence to working better and faster. The LCMS-triple-quadrupole combines all UF technologiesand pushes the limits of LC-MS/MS quantitationfor applications requiring highest sensitivity androbustness.

World’s highest sensitivitybased on the new UF Qarray technology, deliver -ing new limits of MRM sensitivity and impactingfull-scan sensitivity

Unmatched speeddue to data acquisition with scan speed of 30,000 u/sec and polarity switching time of 5 msec

Outstanding durabilityachieving peak area response RSD of 3.5 %*, thus showing high robustness

www.shimadzu.eu

*2,400 samples of femtogram levels of alprazolam spiked

into protein-precipitated human plasma extracts over a

6 day period (over 400 samples were injected each day).

Page 44: Exploring the Potential of Continuous Coatingtisztateritermekek.hu/wp-content/uploads/2015/09/Clean... · 2015-11-12 · 18 Exploring the Potential of Continuous Coating Industy experts

Our 20 R&D teams in 10 countries are now

working on 500+ projects, applying multiple

proven and innovative drug delivery technologies

to help you deliver optimal release profi les,

enhanced bioavailability and better dose

forms—preferred by patients and payers.

best technologies. broadest expertise.faster development.

World’s best orally dissolvable tablet – about 3 seconds. Uniquely delivers small or large molecules. 20 Rx and OTC products in 50 markets. NEW taste-masking and higher dose options.

technology

spotlight

rp scherer softgel technologies

controlled release technologies

optimelt® hme technology

optidose® technology

zydis® fast dissolve technologies

˝ 2

015

Cata

len

t P

harm

a S

olu

tio

ns.

All r

igh

ts r

ese

rve

d.

Catalent. More products. Better treatments. Reliably supplied.™

us + 1 888 SOLUTION eu + 800 8855 6178 [email protected] catalent.com

TECHNOLOGY SELECTION & APPLICATION � FORMULATION & ANALYTICAL SERVICES � CLINICAL & COMMERCIAL SUPPLY ��TAILORED OR END-TO-END SOLUTIONS