throbber
PHARMACEUTICAL
`MANUFACTURING
`HANDBOOK
`Production and
`Processes
`
`SHAYNE COX GAD, PH.D., D.A.B.T.
`Gad Consulting Services
`Cary, North Carolina
`
`A JOHN WILEY & SONS, INC., PUBLICATION
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 1 of 102
`
`

`

`Copyright © 2008 by John Wiley & Sons, Inc. All rights reserved
`
`Published by John Wiley & Sons, Inc., Hoboken, New Jersey
`Published simultaneously in Canada
`
`No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any
`form or by any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except
`as permitted under Section 107 or 108 of the 1976 United States Copyright Act, without either the
`prior written permission of the Publisher, or authorization through payment of the appropriate per-
`copy fee to the Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, (978)
`750-8400, fax (978) 750-4470, or on the web at www.copyright.com. Requests to the Publisher for per-
`mission should be addressed to the Permissions Department, John Wiley & Sons, Inc., 111 River
`Street, Hoboken,
`NJ 07030, (201) 748-6011, fax (201) 748-6008, or online at http://www.wiley.com/go/permission.
`
`Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts
`in preparing this book, they make no representations or warranties with respect to the accuracy
`or completeness of contents of this book and specifi cally disclaim any implied warranties of
`merchantability or fi tness for a particular purpose. No warranty may be created or extended by sales
`representatives or written sales materials. The advice and strategies contained herein may not be
`suitable for your situation. You should consult with a professional where appropriate. Neither the
`publisher nor author shall be liable for any loss of profi t or any other commercial damages, including
`but not limited to special, incidental, consequential, or other damages.
`
`For general information on our other products and services or for technical support, please contact
`our Customer Care Department within the United States at (800) 762-2974, outside the United States
`at (317) 572-3993 or fax (317) 572-4002.
`
`Wiley also publishes its books in a variety of electronic formats. Some content that appears in print
`may not be available in electronic formats. For more information about Wiley products, visit our web
`site at www.wiley.com.
`
`Library of Congress Cataloging-in-Publication Data is available.
`
`ISBN: 978-0-470-25958-0
`
`Printed in the United States of America
`
`10 9 8 7 6 5 4 3 2 1
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 2 of 102
`
`

`

`CONTENTS
`
`PREFACE
`
`SECTION 1 MANUFACTURING SPECIALTIES
`
`1.1 Biotechnology-Derived Drug Product Development
`Stephen M. Carl, David J. Lindley, Gregory T. Knipp, Kenneth R. Morris,
`Erin Oliver, Gerald W. Becker, and Robert D. Arnold
`
`1.2 Regulatory Considerations in Approval on Follow-On Protein
`Drug Products
`Erin Oliver, Stephen M. Carl, Kenneth R. Morris, Gerald W. Becker, and
`Gregory T. Knipp
`
`1.3 Radiopharmaceutical Manufacturing
`Brit S. Farstad and Iván Peñuelas
`
`SECTION 2 ASEPTIC PROCESSING
`
`2.1
`
`Sterile Product Manufacturing
`James Agalloco and James Akers
`
`SECTION 3 FACILITY
`
`3.1
`
`From Pilot Plant to Manufacturing: Effect of Scale-Up on
`Operation of Jacketed Reactors
`B. Wayne Bequette
`
`xiii
`
`1
`
`3
`
`33
`
`59
`
`97
`
`99
`
`137
`
`139
`
`ix
`IPR2020-00770
`United Therapeutics EX2018
`Page 3 of 102
`
`

`

`x
`
` CONTENTS
`
`3.2 Packaging and Labeling
`Maria Inês Rocha Miritello Santoro and Anil Kumar Singh
`
`3.3 Clean-Facility Design, Construction, and Maintenance Issues
`Raymond K. Schneider
`
`SECTION 4 NORMAL DOSAGE FORMS
`
`4.1
`
`4.2
`
`Solid Dosage Forms
`Barbara R. Conway
`
`Semisolid Dosages: Ointments, Creams, and Gels
`Ravichandran Mahalingam, Xiaoling Li, and Bhaskara R. Jasti
`
`4.3 Liquid Dosage Forms
`Maria V. Rubio-Bonilla, Roberto Londono, and Arcesio Rubio
`
`SECTION 5 NEW DOSAGE FORMS
`
`5.1 Controlled-Release Dosage Forms
`
`Anil Kumar Anal
`
`5.2 Progress in the Design of Biodegradable Polymer-Based
`Microspheres for Parenteral Controlled Delivery of Therapeutic
`Peptide/Protein
`Shunmugaperumal Tamilvanan
`
`
`
`5.3 Liposomes and Drug Delivery
`Sophia G. Antimisiaris, Paraskevi Kallinteri, and Dimitrios G. Fatouros
`
`5.4 Biodegradable Nanoparticles
`Sudhir S. Chakravarthi and Dennis H. Robinson
`
`5.5 Recombinant Saccharomyces cerevisiae as New Drug Delivery
`System to Gut: In Vitro Validation and Oral Formulation
`Stéphanie Blanquet and Monique Alric
`
`5.6 Nasal Delivery of Peptide and Nonpeptide Drugs
`Chandan Thomas and Fakhrul Ahsan
`
`5.7 Nasal Powder Drug Delivery
`(cid:252)
`(cid:254) (cid:252)
`
`Jelena Filipovi -Gr i and Anita Hafner
`
`5.8 Aerosol Drug Delivery
`
`Michael Hindle
`
`5.9 Ocular Drug Delivery
`Ilva D. Rupenthal and Raid G. Alany
`
`5.10 Microemulsions as Drug Delivery Systems
`Raid G. Alany and Jingyuan Wen
`
`159
`
`201
`
`233
`
`235
`
`267
`
`313
`
`345
`
`347
`
`393
`
`443
`
`535
`
`565
`
`591
`
`651
`
`683
`
`729
`
`769
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 4 of 102
`
`

`

`CONTENTS
`
` xi
`
`5.11 Transdermal Drug Delivery
`C. Scott Asbill and Gary W. Bumgarner
`
`5.12 Vaginal Drug Delivery
`José das Neves, Maria Helena Amaral, and Maria Fernanda Bahia
`
`SECTION 6 TABLET PRODUCTION
`
`6.1 Pharmaceutical Preformulation: Physicochemical Properties of
`Excipients and Powers and Tablet Characterization
`Beom-Jin Lee
`
`
`
`6.2 Role of Preformulation in Development of Solid Dosage Forms
`Omathanu P. Perumal and Satheesh K. Podaralla
`
`6.3 Tablet Design
`Eddy Castellanos Gil, Isidoro Caraballo, and Bernard Bataille
`
`6.4 Tablet Production Systems
`Katharina M. Picker-Freyer
`
`6.5 Controlled Release of Drugs from Tablet Coatings
`Sacide Alsoy Altinkaya
`
`6.6 Tablet Compression
`Helton M. M. Santos and João J. M. S. Sousa
`
`6.7 Effects of Grinding in Pharmaceutical Tablet Production
`Gavin Andrews, David Jones, Hui Zhai, Osama Abu Diak, and
`Gavin Walker
`
`6.8 Oral Extended-Release Formulations
`Anette Larsson, Susanna Abrahmsén-Alami, and Anne Juppo
`
`SECTION 7 ROLE OF NANOTECHNOLOGY
`
`7.1 Cyclodextrin-Based Nanomaterials in Pharmaceutical Field
`Erem Bilensoy and A. Attila Hincal
`
`7.2 Nanotechnology in Pharmaceutical Manufacturing
`
`Yiguang Jin
`
`7.3 Pharmaceutical Nanosystems: Manufacture, Characterization,
`and Safety
`D. F. Chowdhury
`
`7.4 Oil-in-Water Nanosized Emulsions: Medical Applications
`
`Shunmugaperumal Tamilvanan
`
`INDEX
`
`793
`
`809
`
`879
`
`881
`
`933
`
`977
`
`1053
`
`1099
`
`1133
`
`1165
`
`1191
`
`1223
`
`1225
`
`1249
`
`1289
`
`1327
`
`1367
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 5 of 102
`
`

`

`1.1
`
` BIOTECHNOLOGY - DERIVED DRUG
`PRODUCT DEVELOPMENT
`
` Stephen M. Carl, 1 David J. Lindley, 1 Gregory T. Knipp, 1
` Kenneth R. Morris, 1 Erin Oliver, 2 Gerald W. Becker, 3 and
` Robert D. Arnold 4
`1 Purdue University, West Lafayette, Indiana
`2 Rutgers, The State University of New Jersey, Piscataway, New Jersey
`3 SSCI, West Lafayette, Indiana
`4 The University of Georgia, Athens, Georgia
`
` Contents
`
` 1.1.3
`
` 1.1.4
`
` 1.1.1 Introduction
` 1.1.2
` Formulation Assessment
` 1.1.2.1
` Route of Administration and Dosage
` 1.1.2.2
` Pharmacokinetic Implications to Dosage Form Design
` 1.1.2.3
` Controlled - Release Delivery Systems
` Analytical Method Development
` 1.1.3.1
` Traditional and Biophysical Analytical Methodologies
` 1.1.3.2
` Stability - Indicating Methodologies
` 1.1.3.3
` Method Validation and Transfer
` Formulation Development
` 1.1.4.1
` Processing Materials and Equipment
` 1.1.4.2
` Container Closure Systems
` 1.1.4.3
` Sterility Assurance
` 1.1.4.4
` Excipient Selection
` Drug Product Stability
` 1.1.5.1
` Defi ning Drug Product Storage Conditions
` 1.1.5.2
` Mechanisms of Protein and Peptide Degradation
` 1.1.5.3
` Photostability
` 1.1.5.4
` Mechanical Stress
` 1.1.5.5
` Freeze – Thaw Considerations and Cryopreservation
` 1.1.5.6
` Use Studies
` 1.1.5.7
` Container Closure Integrity and Microbiological Assessment
` 1.1.5.8
` Data Interpretation and Assessment
`
` 1.1.5
`
`Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad
`Copyright © 2008 John Wiley & Sons, Inc.
`
`3
`IPR2020-00770
`United Therapeutics EX2018
`Page 6 of 102
`
`

`

`4
`
` 1.1.6
`
` 1.1.7
`
`BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT
`
` Quality by Design and Scale - Up
` 1.1.6.1
` Unit Operations
` 1.1.6.2
` Bioburden Considerations
` 1.1.6.3
` Scale - Up and Process Changes
` Concluding Remarks
` References
`
` 1.1.1
`
` INTRODUCTION
`
` Although the origins of the fi rst biological and/or protein therapeutics can be
`traced to insulin in 1922, the fi rst biotechnology - derived pharmaceutical drug
`product approved in the United States was Humulin in 1982. In the early stages
`of pharmaceutical biotechnology, companies that specialized primarily in the devel-
`opment of biologicals were the greatest source of research and development in
`this area. Recent advances in molecular and cellular biological techniques and
`the potential clinical benefi ts of biotechnology drug products have led to a sub-
`stantial increase in their development by biotechnology and traditional pharma-
`ceutical companies . In terms of pharmaceuticals, the International Conference on
`Harmonization (ICH) loosely defi nes biotechnology - derived products with biologi-
`cal origin products as those that are “ well - characterized proteins and polypeptides,
`their derivatives and products of which they are components, and which are
`isolated from tissues, body fl uids, cell cultures, or produced using rDNA tech-
`nology ” [1] . In practical terms, biological and biotechnology - derived pharmaceuti-
`cal agents encompass a number of therapeutic classes, including cytokines,
`erythropoietins, plasminogen activators, blood plasma factors, growth hormones
`and growth factors, insulins, monoclonal antibodies, and vaccines [1] . Additionally,
`short interfering and short hairpin ribonucleic acids (siRNA, shRNA) and anti-
`sense oligonucleotide therapies are generally characterized as biotechnology -
` derived products.
` According to the biotechnology advocacy group, The Biotechnology Industry
`Organization (BIO), pharmaceutical - based biotechnology represents over a $ 30
`billion dollar a year industry and is directly responsible for the production of
`greater than 160 drug therapeutics and vaccines [2] . Furthermore, there are more
`than 370 biotechnology - derived drug products and vaccines currently in clinical
`trials around the world, targeting more than 200 diseases, including various cancers,
`Alzheimer ’ s disease, heart disease, diabetes, multiple sclerosis, acquired immuno-
`defi ciency syndrome (AIDS), and arthritis. While the clinical value of these
`products is well recognized, a far greater number of biotechnology - derived drug
`products with therapeutic potential for life - altering diseases have failed in
`development.
` As the appreciation of the clinical importance and commercial potential for bio-
`logical products grows, new challenges are arising based on the many technological
`limitations related to the development and marketing of these complex agents.
`Additionally, the intellectual property protection of an associated agent might not
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 7 of 102
`
`

`

`INTRODUCTION
`
`5
`
`provide a suffi cient window to market and regain the costs associated with the dis-
`covery, research, development, and scale - up of these products. Therefore, to prop-
`erly estimate the potential return on investment, a clear assessment of potential
`therapeutic advantages and disadvantages, such as the technological limitations in
`the rigorous characterization required of these complex therapeutic agents to gain
`Food and Drug Administration (FDA) approval, is needed prior to initiating
`research. Clearly, research focused on developing methodologies to minimize these
`technological limitations is needed. In doing so we hypothesize the attrition rate
`can be reduced and the number of companies engaged in the development of bio-
`technology - derived products and diversity of products will continue to expand.
` Technological limitations have limited the development of follow - on, or generic
`biopharmaceutical products that have lost patent protection. In fact, the potential
`pitfalls associated with developing these compounds are so diverse that regulatory
`guidance concerning follow - on biologics is relatively obscure and essentially notes
`that products will be assessed on a case - by - case basis. The reader is encouraged to
`see Chapter 1.2 for a more detailed discussion concerning regulatory perspectives
`pertaining to follow - on biologics.
` Many of the greatest challenges in producing biotechnology - derived pharmaceu-
`ticals are encountered in evaluating and validating the chemical and physical nature
`of the host expression system and the subsequent active pharmaceutical ingredient
`(API) as they are transferred from discovery through to the development and mar-
`keting stages. Although this area is currently a hotbed of research and is progressing
`steadily, limitations in analytical technologies are responsible for a high degree of
`attrition of these compounds. The problem is primarily associated with limited
`resolution of the analytical technologies utilized for product characterization. For
`example, without the ability to resolve small differences in secondary or tertiary
`structure, linking changes to product performance or clinical response is impossible.
`The biological activity of traditional small molecules is related directly to their
`structure and can be determined readily by nuclear magnetic resonance (NMR),
`X - Ray crystallography (X - ray), mass spectrometry (MS), and/or a combination of
`other spectroscopic techniques. However, methodologies utilized for characterizing
`biological agents are limited by resolution and reproducibility. For instance, circular
`dichroism (CD) is generally considered a good method to determine secondary
`structural elements and provides some information on the folding patterns (tertiary
`structure) of proteins. However, CD suffers from several limitations, including a
`lower resolution that is due in part to the sequence libraries used to deconvolute
`the spectra. To improve the reliability of determining the secondary and tertiary
`structural elements, these databases need to be developed further. An additional
`example is the utility of two - dimensional NMR (2D - NMR) for structural determi-
`nation. While combining homonuclear and heteronuclear experimental techniques
`can prove useful in structural determination, there are challenges in that 2D - NMR
`for a protein could potentially generate thousands of signals. The ability to assign
`specifi c signals to each atom and their respective interactions is a daunting task.
`Resolution between the different amino acids in the primary sequence and their
`positioning in the covalent and folded structures become limited with increasing
`molecular weight. Higher dimensional techniques can be used to improve resolu-
`tion; however, the resolution of these methods remains limited as the number of
`amino acids is increased.
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 8 of 102
`
`

`

`6
`
`BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT
`
` Understanding the limitations of the analytical methodologies utilized for product
`characterization has led to the development of new experimental techniques as well
`as the refi ned application of well - established techniques to this emerging fi eld. Only
`through application of a number of complementary techniques will development
`scientists be able to accurately characterize and develop clinically useful products.
`Unfortunately, much of the technology is still in its infancy and does not allow for
`a more in - depth understanding of the subtleties of peptide and protein processing
`and manufacturing. For instance, many of the analytical techniques utilized for
`characterization will evaluate changes to product conformation on the macroscopic
`level, such as potential denaturation or changes in folding, as observed with CD.
`However, these techniques do not afford the resolution to identify subtle changes
`in conformation that may either induce chemical or physical instabilities or unmask
`antigenic epitopes.
` Further limiting successful product development is a lack of basic understanding
`as to critical manufacturing processes that have the potential to affect the structural
`integrity and activity of biopharmaceuticals. As with traditional small molecules,
`stresses associated with the different unit operations may affect biopharmaceutical
`products differently. In contrast to traditional small molecules, there is considerable
`diffi culty in identifying potentially adverse affects, if any, that a particular unit opera-
`tion may have on the clinically critical structural elements of a drug. Considering
`that many proteins exhibit a greater potential for degradation from shear stress, it
`is particularly important to assess any negative effects of mixing, transport through
`tubing, fi ltration, and fi lling operations. Essentially all unit operations for a given
`manufacturing process could create enough shear stress to induce minor structural
`changes that could lead to product failure. The diffi culty is establishing what degree
`of change will have an impact on the stability, bioactivity, or immunogenic potential
`of the compound. Unfortunately, unless exhaustive formulation development studies
`are conducted, coupled with a comprehensive spectrum of analytical methodologies,
`these effects may not be readily evident until after scale - up of the manufacturing
`process or, worse yet, in the clinical setting. Moreover, modeling shear and stress
`using fl uid dynamic structurally diverse molecules is a foreboding task. Extending
`these models to validate process analytical technologies (PAT) and incorporate
`critical quality by design (QbD) elements in the development process for a collec-
`tion of biopharmaceuticals would be largely hindered by the daunting nature of the
`task at hand.
` The use of biological systems to produce these agents results in additional vari-
`ability. Slight changes in nutrient profi le could affect growth patterns and protein
`expression of cultured cells. Furthermore, microbial contamination in the form of
`viruses, bacteria, fungi, and mycoplasma can be introduced during establishment of
`cell lines, cell culture/fermentation, capture and downstream processing steps, for-
`mulation and fi lling operations, or drug delivery [3] . Therefore, establishing the
`useful life span of purifi cation media and separation columns remains a critical issue
`for consistently producing intermediates and fi nal products that meet the defi ned
`quality and safety attributes of the product [4] . In short, understanding the proper
`processability and manufacturing controls needed has been a major hurdle that has
`kept broader development of biopharmaceutical products relatively limited.
` Notwithstanding the many technological hurdles to successfully develop a phar-
`maceutically active biotechnology product, they offer many advantages in terms of
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 9 of 102
`
`

`

`FORMULATION ASSESSMENT
`
`7
`
`therapeutic potency, specifi city, and target design (not generally limited to a particu-
`lar class or series of compounds). This is an iterative approach, whereby every new
`approved compound, new lessons, and applications to ensure successful product
`development are realized, thereby adding to our knowledge base and facilitating
`the development of future products. This chapter will discuss some of the funda-
`mental issues associated with successful biopharmaceutical drug product develop-
`ment and aims to provide an understanding of the subtleties associated with their
`characterization, processing, and manufacturing.
`
` 1.1.2
`
` FORMULATION ASSESSMENT
`
` In order to select the most appropriate formulation and route of administration for
`a drug product, one must fi rst assess the properties of the API, the proposed thera-
`peutic indication, and the requirements/limitations of the drug and the target patient
`population. Development teams are interdisciplinary comprised of individuals with
`broad expertise, for example, chemistry, biochemistry, bioengineering, and pharma-
`ceutics, that can provide insight into the challenges facing successful product devel-
`opment. As such, knowledge gained through refi nement of the API manufacturing
`process through to lead optimization is vital to providing an initial starting point
`for success. Information acquired, for example, in the way of analytical development
`and API characterization, during drug discovery or early preclinical development
`that can be applied to fi nal drug product development may contribute to shorter
`development times of successful products.
` The host system utilized for API production is critical to the production of the
`fi nal product and will determine the basic and higher order physicochemical char-
`acteristics of the drug. Typically biopharmaceuticals are manufactured in Escherichia
`coli as prokaryotic and yeast and Chinese hamster ovary (CHO) cells as eukaryotic
`expression systems [5] . While general procedures for growth condition optimization
`and processing and purifi cation paradigms have emerged, differences in posttrans-
`lational modifi cations and host – system related impurities can exist even with rela-
`tively minor processing changes within a single production cell line [5] . Such changes
`have the potential to alter the biopharmaceutical properties of the active compound,
`its bioactivity , and its potential to elicit adverse events such as immunogenic reac-
`tions. These properties will be a common theme as they could potentially play a
`major role in both analytical and formulation development activities.
` During the process of lead optimization, characterization work is performed that
`would include a number of parameters that are critical to formulation and analytical
`development scientists. The following information is a minimalist look at what
`information should be available to support product development scientists:
`
` • Color
` • Particle size and morphology (for solid isolates)
` • Thermoanalytical profi le (e.g., Tg for lyophiles)
` • Hygroscopicity
` • Solubility with respect to pH
` • Apparent solution pH
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 10 of 102
`
`

`

`8
`
`BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT
`
`䊊
`
`䊊
`
`䊊
`
`䊊
`
` • Number and p Ka of ionizable groups
` • Amino acid sequence
` • Secondary and tertiary structural characteristics
` • Some stability parameters with respect to
`䊊 pH
` Temperature
` Humidity
` Light
` Mechanical stress
` Oxygen sensitivity
`• Impurity profi le
`䊊 Misfolded/misaligned active
` Potential isoforms
` Expression system impurities
`• Potency [median inhibitory concentration (IC 50 )]
`• Animal Pharmacokinetic/Pharmacodynamic (PK/PD) and Tox profi les
`
`䊊
`
`䊊
`
`䊊
`
` All of the above information will prove invaluable in determining the potential
`methods for rational drug delivery. Particular attention should be paid to the rela-
`tive hygroscopicity of the API, of course, any stability information, as well as the
`impurity profi le and ADMET (absorption, distribution, metabolism, excretion, and
`toxicity) information. In short, the more information that is available when develop-
`ment activities are initiated, the easier it is to avoid common pitfalls and make
`development decisions more rationally.
`
` 1.1.2.1
`
` Route of Administration and Dosage
`
` Biologics are traditionally very potent molecules that may require only picomolar
`blood concentrations to elicit a therapeutic effect. Given that the amount of drug
`required per dosage will be commensurate with the relative potency of the molecule,
`small concentrations are generally required for any unit dose. Biopharmaceuticals
`typically have large molecular weights relative to conventional pharmaceutical
`agents, which may be increased further by posttranslational modifi cations. The phar-
`macokinetics (ADMET) of biotechnology products have been reviewed elsewhere
` [6] , but generally they have short circulating half - lives [7] . As such, biological prod-
`ucts are most often delivered parenterally and formulated as solutions, suspensions,
`or lyophilized products for reconstitution [8, 9] . However, one must fi rst ascertain
`the potential physiological barriers to drug delivery and effi cacy before assessing
`potential routes of administration. These barriers may include actual physical bar-
`riers, such as a cell membrane, that could restrict the drug from reaching its site of
`action or chemical barriers, including pH or enzymatic degradation. Based on
`current drug delivery approaches, the proteinaceous nature of biological products
`limits their peroral delivery due to their susceptibility to proteases and peptidases
`present in the gastrointestinal tract as well as size limitations for permeating through
`absorptive enterocytes [10] .
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 11 of 102
`
`

`

`FORMULATION ASSESSMENT
`
`9
`
` Diffi culties in peroral delivery have stimulated researchers to explore alternate
`delivery mechanisms for biologics, such as through the lungs or nasal mucosa [11,
`12] . Further, advances in technology and our understanding of the mechanisms
`limiting oral delivery of biotechnology products have led to innovative drug delivery
`approaches to achieve suffi cient oral bioavailability. However, no viable products
`have successfully reached the market [13] . As a result of the technological limita-
`tions inherent in biopharmaceutical delivery, these compounds are largely delivered
`parenterally through an injection or implant.
` When assessing the potential routes of administration, one must consider the
`physicochemical properties of the drug, its ADMET properties, the therapeutic
`indication, and the patient population, some of which are discussed below. Table 1
`provides a list of some of those factors that must be addressed when determining
`the most favorable route of administration and the subsequent formulation for
`delivery. Ideally the route of administration and subsequent formulation will be
`optimized after identifying critical design parameters to satisfy the needs of patients
`and health care professionals alike while maintaining the safety and effi cacy of the
`product.
` Parenteral administration is the primary route of delivering biopharmaceutical
`agents (e.g., insulin); however, issues associated with patient compliance with admin-
`istration of short - acting molecules are a challenge. Yet, the risk - to - benefi t ratio must
`be weighed when determining such fundamental characteristics of the fi nal dosage
`form. For instance, a number of biopharmaceutical compounds are administered
`subcutaneously, but this route of parenteral administration exhibits the highest
`potential for immunogenic adverse events due to the presence of Langerhans cells
` [14] . A compound ’ s immunogenic potential is related to a host of factors, both
`
` Factors That Determine Route of
` TABLE 1
`Administration
`
` Site of action
` Therapeutic indication
` Dosage
` Potency/biological activity
` Pharmacokinetic profi le
`
` Absorption time from tissue vs. IV
`
` Circulating half - life
`
` Distribution and elimination kinetics
` Toxicological profi le
` Immunogenic potential
` Patient population characteristics
`
` Disease state
`
` Pathophysiology
`
` Age
` Pharmacodynamic profi le
`
` Onset and duration of action
`
` Required clinical effect
` Formulation considerations
`
` Stability
`
` Impurity profi le
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 12 of 102
`
`

`

`10
`
`BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT
`
`patient and treatment related; however, if an alternate, potentially safer route of
`administration is available, it may be prudent to consider it. Other factors, such as
`the frequency of dosing (especially into an immune organ such as the skin) and the
`duration of treatment, can also dramatically increase the potential for immunogenic
`reactions [14] . Many of the factors that contribute to the immunogenic potential of
`biopharmaceuticals, such as impurities, degradation products, and native antigenic
`epitopes, can be mitigated through altering the physicochemical properties of the
`drug (e.g., pegylation [15, 16] , acylation [17, 18] , increased glycosylation to mask
`epitopes [19] ) or changing the characteristics of the formulation [20, 21] . In reality,
`the pharmaceutical industry has done a good job of recognizing the potential impli-
`cations of immunogenic reactions and readily embraced technologies that can either
`mask or eliminate potential antigenic epitopes. However, additional research is
`needed to further identify and remove immunogenic epitopes.
`
` 1.1.2.2
`
` Pharmacokinetic Implications to Dosage Form Design
`
` Biological agents are generally eliminated by metabolism into di - and tripeptides,
`amino acids, and smaller components for subsequent absorption as nutrients or
`clearance by the kidney, liver, or other routes. Renal elimination of peptides and
`proteins occur primarily via three distinct mechanisms. The fi rst involves the glo-
`merular fi ltration of low - molecular - weight proteins followed by reabsorption into
`endocytic vesicles in the proximal tubule and subsequent hydroysis into small peptide
`fragments and amino acids [22] . Interleukin 11 (IL - 11) [23] , IL - 2 [24] , insulin [25] ,
`and growth hormone [26] have been shown to be eliminated by this method. The
`second involves hydrolysis of the compound at the brush border of the lumen and
`subsequent reabsorption of the resulting metabolites [6] . This route of elimination
`applies to small linear peptides such as angiotensin I and II, bradykinin, glucagons,
`and leutinizing hormone releasing hormone (LHRH) [6, 27, 28] . The third route of
`renal elimination involves peritubular extraction from postglomerular capillaries
`and intracellular metabolism [6] . Hepatic elimination may also play a major role in
`the metabolism of peptides and proteins; however, reticuloendothelial elimination is
`by far the primary elimination route for large macromolecular compounds [29] .
` Biopharmaceutical drug products are subject to the same principles of pharma-
`cokinetics and exposure/response correlations as conventional small molecules [6] .
`However, these products are subject to numerous pitfalls due to their similarity to
`nutrients and endogenous proteins and the evolutionary mechanisms to break them
`down or prevent absorption. The types of pharmacokinetic - related problems that a
`biotechnology drug development team may encounter range from lack of specifi city
`and sensitivity of bioanalytical assays to low bioavailability and rapid drug elimina-
`tion from the system [6] . For example, most peptides have hormone activity and
`usually short elimination half - lives which can be desirable for close regulation of
`their endogenous levels and function. On the other hand, some proteins such as
`albumin or antibodies have half - lives of several days and formulation strategies
`must be designed to account for these extended elimination times [6] . For example,
`the reported terminal half - life for SB209763, a humanized monoclonal antibody
`against respiratory syncytial virus, was reported as 22 – 50 days [30] . Furthermore,
`some peptide and protein products that persist in the bloodstream exhibit the
`potential for idiosyncratic adverse affects as well as increased immunogenic poten-
`
`IPR2020-00770
`United Therapeutics EX2018
`Page 13 of 102
`
`

`

`ANALYTICAL METHOD DEVELOPMENT
`
`11
`
`tial. Therefore, the indication and formulation strategy can prove crucial design
`parameters simply based on clearance mechanisms.
`
` 1.1.2.3
`
` Controlled - Release Delivery Systems
`
` Given that the majority of biopharmaceutical products are indicated for chronic
`conditions and may require repeated administrations, products may be amenable to
`controlled - release drug delivery systems. Examples include Lupron Depot (leupro-
`lide acetate), which is delivered subcutaneously in microspheres [31] , and Viadur,
`which is implanted subcutaneous

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket