throbber
Practical Process Research
`and Development
`A guide for organic chemists
`
`Second Edition
`
`Neal G. Anderson
`Anderson’s Process Solutions
`Jacksonville, Oregon
`
`AMSTERDAM l BOSTON l HEIDELBERG l LONDON
`NEW YORK l OXFORD l PARIS l SAN DIEGO
`SAN FRANCISCO l SYDNEY l TOKYO
`
`Academic Press is an imprint of Elsevier
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 1 of 86
`
`

`

`Academic Press is an imprint of Elsevier
`The Boulevard, Langford Lane, Kidlington, Oxford, OX5 1GB, UK
`225 Wyman Street, Waltham, MA 02451, USA
`Radarweg 29, PO Box 211, 1000 AE Amsterdam, The Netherlands
`Copyright Ó 2012 Elsevier Inc. All rights reserved.
`
`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 or otherwise without
`the prior written permission of the publisher
`
`Permissions may be sought directly from Elsevier’s Science & Technology Rights Department in
`Oxford, UK: phone (+44) (0) 1865 843830; fax (+44) (0) 1865 853333; email: permissions@
`elsevier.com. Alternatively you can submit your request online by visiting the Elsevier web site at
`http://elsevier.com/locate/permissions, and selecting Obtaining permission to use Elsevier material
`
`Notice
`No responsibility is assumed by the publisher for any injury and/or damage to persons or property
`as a matter of products liability, negligence or otherwise, or from any use or operation of any
`methods, products, instructions or ideas contained in the material herein
`
`British Library Cataloguing in Publication Data
`A catalogue record for this book is available from the British Library
`
`Library of Congress Cataloging-in-Publication Data
`Anderson, Neal G.
`Practical process research and development : a guide for organic
`chemists / Neal G. Anderson. – 2nd ed.
`p. cm.
`Rev. ed. of: Practical process research & development. c2000.
`ISBN 978-0-12-386537-3 (hardback)
`1. Chemical processes. I. Anderson, Neal G. Practical process
`research & development. II. Title. III. Title: Practical process
`research and development.
`TP155.7.A55 2000
`541’.39–dc23
`
`2011051049
`
`ISBN: 978-0-12-386537-3
`
`For information on all Academic Press publications
`visit our website at books.elsevier.com
`
`Printed and bound in the USA
`12 13 14 15 16 10 9 8 7 6 5 4 3 2 1
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 2 of 86
`
`

`

`Chapter 1
`
`Introduction
`
`Chapter Outline
`I. Introduction
`II. Equipment Considerations
`on Scale
`III. Operations Preferred
`on Scale
`
`1
`
`7
`
`12
`
`IV. Patent Considerations
`V. Summary and Perspective
`References
`
`15
`16
`19
`
`“The world doesn’t move because of idealism.. [i]t moves because of economic incentives.”
`– Fernando Canales Clariond, formerly Mexico’s secretary of the economy [1]
`
`“It is well-known that there are no technical optima in industry, only economic optima..”
`– G. Guichon et al. [2]
`
`“Today, green chemistry is simply a good business choice.”
`
`– Paul Anastas [3]
`
`I. INTRODUCTION
`
`The driving forces of the pharmaceutical industry are to develop medicines that maintain
`or improve health and the quality of life for people, and to provide a reasonable return for
`investors. The many unknowns of the business, especially our imperfect understanding
`of biology, make for a high-risk environment. Yet the rewards are high as well. In
`Table 1.1 are presented some statistics associated with developing drugs, and these
`statistics explain some of the pressures of the business.
`Significant financial gains are possible by developing drugs, as indicated by the
`penalties and fines levied against firms and people recently. For instance, in 2010
`AstraZeneca was fined $520,000,000 for promoting Seroquel for off-label uses [4] and
`GlaxoSmithKline was fined $150,000,000 for violations of current Good Manufacturing
`Practices (cGMPs) [5]. In 2009, Pfizer was fined $2,300,000,000 for illegally promoting
`
`Practical Process Research and Development. DOI: 10.1016/B978-0-12-386537-3.00001-0
`Copyright Ó 2012 Elsevier Inc. All rights reserved.
`
`1
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 3 of 86
`
`

`

`2
`
`Practical Process Research and Development
`
`TABLE 1.1 Some Statistics Relevant to the Pharmaceutical Industry
`
`Value
`
`Factor
`
`$1,300,000,000
`
`Cost to bring a drug to market (1)
`
`5e20% of drug product price
`
`CoG of API
`
`30% of the CoG for
`drug product
`
`Cost of QC (2)
`
`Millions of dollars
`
`Cost of failed drug formulation (3)
`
`As high as market will bear
`
`Price of drug product to consumer
`(higher for US than for most countries) (4)
`
`$75,000
`
`The value of one additional year of life, set in 2005 by
`health economists (5)
`
`$200,000e$300,000
`
`Annual cost of US chemist or engineer for an employer
`
`About 95%
`
`About 30%
`
`8 years
`
`20 years
`
`Portion of drug candidates that fail in pre-clinical or clinical
`studies
`
`Portion of approved drugs that recoup development costs (6)
`
`Average time of development (goal is 5 years)
`
`Period for exclusive sales of a patented drug (US)
`
`20 e Development time
`
`Years to recoup investment costs
`
`$1,000,000
`
`Sales lost for every day that a filing is delayed, if drug sales
`are $400 MM/year
`
`(1) Undoubtedly includes the cost of advancing drug candidates that failed. Jarvis, L. M. Chem. Eng. News 2010,
`88(23), 13. May be higher: Vertex developed Incivek (telaprevir) over 20 years at the cost of around
`$4,000,000,000: Jarvis, L. Chem. Eng. News 2011, 89(22), 8.
`(2) Mullin, R. Chem. Eng. News 2009, 87(39), 38.
`(3) May be higher for formulations involving spray drying. A huge cost may be incurred if the physical form of an
`API is not controlled and a dosage form fails specifications, thus interrupting clinical trials or discontinuing
`sales of a drug (Chapter 13). Thayer, A. M. Chem. Eng. News 2010, 88(22), 13.
`(4) http://www.economist.com/node/4054095 (June 16, 2005).
`(5) The Washington Post, July 17, 2005: http://www.washingtonpost.com/wp-dyn/content/article/2005/07/16/
`AR2005071600941_pf.html.
`(6) Mod. Drug Discov. 2001, 4(10), 47.
`
`Bextra and three other medications [6], and Bristol–Myers Squibb (BMS) was fined
`$2,100,000 for making agreements with Apotex to delay the launch of generic Plavix
`[7]. In 2009, the FDA stopped reviewing applications from Ranbaxy and prohibited
`Ranbaxy from importing 30 generic drugs, due to falsified QC data [8]. In 2007, a former
`top official of China’s state organization approving drugs was executed for taking bribes
`[9]. In 2004, BMS was fined $150,000,000 for “channel stuffing,” an accounting practice
`that artificially boosted the sale of drugs [10]. The reputation of the pharmaceutical
`industry has been sullied over the past few decades, but many people, including this
`author, enter this industry because they want to be able to help others.
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 4 of 86
`
`

`

`Chapter | 1 Introduction
`
`3
`
`The development and sales of drugs are influenced by an interplay of financial,
`political, governmental and personal considerations. For example, Bayer reduced the
`cost of a pill of ciprofloxacin from $1.77 to $0.75 after the horrific crashes of
`September 11, 2001 [11]. As of August 2009 the pharmaceutical lobby had 1544
`lobbyists, or almost three lobbyists per congressman in Washington DC [12]; this is
`a sizeable increase from the 625 registered lobbyists in 2001 [11]. No major US
`pharmaceutical company would develop RU-486, an abortifacient, due to backlash
`anticipated from conservative groups. The sale of anti-AIDS drugs at reduced prices to
`the third world is a nice example of philanthropy; probably some income tax write-offs
`are also involved. Philanthropic efforts exist, such as Merck’s gifts of ivermectin to
`prevent river blindness and the development of tenofovir by the Clinton Health Access
`Initiative to treat AIDS in the developing world [13]. Pharmaceutical industries may
`need support from government to continue developing drugs for the third world, such
`as compounds to treat malaria [14]. Efforts to minimize wastes and decrease impacts
`on the environment have increased, due to sensible and altruistic reasons, and due to
`penalties imposed. Although bacteria resistant to powerful antibiotics are continually
`emerging, the development of antibiotics has slowed due to the anticipated longer
`times to recoup development costs from drugs that are not taken on a chronic basis.
`The development of new chemical entities (NCEs) is becoming more difficult with
`increased scrutiny by regulatory authorities; for instance, the third or fourth entry into
`a therapeutic category may have to demonstrate superior benefits to win FDA approval
`[15], and control of potentially genotoxic impurities at the ppm level demands addi-
`tional efforts. And everything is
`influenced by people striving for personal
`advancement.
`As a result of increasing pressure to bring compounds to market, business trends within
`the pharma sector have been changing. Working smarter and faster is stressed, for
`instance, using high-throughput screening and statistically designed experiments. Phar-
`maceutical industries are being pressured to develop more efficient processes [16]. The
`FDA has advanced process analytical
`technology (PAT), and this may decrease
`manufacturing costs [17]. The importance of solid process development efforts has been
`recognized [18].
`The complexity of compounds that has emerged as active pharmaceutical ingredients
`(APIs) may be increasing, as shown in Figure 1.1, and structural complexity increases
`the cost of development. Drug development is expensive: Vertex’s first approved drug
`was developed in-house over 20 years at the cost of around $4,000,000,000. Incivek
`(telaprevir) will treat hepatitis C, at a projected price of $49,200, competing with
`Merck’s Victrelis (boceprevir) at a projected price of $31,000–$44,000. Each company
`has set up assistance programs to help with the co-payments of insured patients [19].
`The foundation of thorough processes is the work carried out by academicians.
`Some brilliant total syntheses have been described by academic chemists, such as the
`syntheses of codeine by the groups of Stork [20] and Magnus [21]. Total synthesis in an
`academic sense [22,23] is often not suitable for scale-up on an industrial scale; scaling
`up in an academic setting to millimoles or grams may not be enough to uncover and
`address processing difficulties. Prof. Hudlicky has described many practical consid-
`erations for syntheses [24] that are applicable to process R&D efforts in the phar-
`maceutical industry.
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 5 of 86
`
`

`

`4
`
`Practical Process Research and Development
`
`CO2Na
`
`O
`
`N
`
`PO O
`
`O
`
`OH
`
`3C CH3
`fosinopril sodium
`
`Ph
`H3C
`
`HS
`
`CH3
`
`N
`
`O
`captopril
`
`CO2H
`
`HN
`
`O
`
`O
`CH3
`
`HN
`
`O
`
`O
`
`N
`
`HN
`
`O
`
`NH
`
`O
`
`N N
`
`F
`
`O
`
`NH2
`
`HN
`
`O
`
`NH
`
`OH
`
`O
`
`HN
`
`O
`
`CH3
`
`H2N
`
`frakefamide
`
`telaprevir
`
`FIGURE 1.1 Structures of APIs and NCE in Table 1.2.
`
`TABLE 1.2 Details on Development of Four APIs and an NCE
`
`Captopril
`(1)
`
`Fosinopril
`(2)
`
`Frakefamide
`(3)
`
`Enfuvirtide
`(4)
`
`Telaprevir
`(5)
`
`Market date
`
`1981
`
`w1988
`
`Halted after
`Phase 2
`
`2003
`
`2011
`
`Steps
`
`Isolations
`
`Preparative
`chromatographies
`
`5
`
`5
`
`0
`
`14
`
`13
`
`0
`
`7
`
`3
`
`0
`
`109
`
`7
`
`2
`
`NA
`
`NA
`
`NA
`
`CoG
`
`NA
`
`NA
`
`$2500/kg
`
`$36,000/kg
`
`NA
`
`Therapeutic area
`
`High blood
`pressure
`
`High blood
`pressure
`
`Pain treatment
`
`AIDS
`
`Hepatitis C
`
`Patient cost/year
`
`NA
`
`NA
`
`NA
`
`$25,000
`
`$49,200
`
`(1) Anderson, N. G.; Bennett, B. J.; Feldman, A. F.; Lust, D. A.; Polomski, R. E. US Patent 5,026,873, 1991
`(to E. R. Squibb & Sons.)
`(2) Grosso, J. A. US Patent 5,162,543, 1992 (to E. R. Squibb & Sons).
`(3) Franze´n, H. M.; Bessidskaia, G.; Abedi, V.; Nilsson, A.; Nilsson, M.; Olsson, L. Org. Process Res. Dev. 2002,
`6, 788.
`(4) Enfuvirtide is a synthetic peptide made of 36 aminoacids: Bray, B. Nat. Rev. Drug Discov. 2003, 2, July, 587.
`(5) Jarvis, L. M. Chem. Eng. News 2011, 89(22), 8.
`
`Perhaps the first description of the principles of process research and development as
`applied to pharmaceutical products was written by Trevor Laird in 1988 [25]; this article
`covered a breadth of topics, including design of experiments (DoEs), automation and the
`importance of polymorphism. Caron recently detailed the responsibilities of the process
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 6 of 86
`
`

`

`Chapter | 1 Introduction
`
`5
`
`chemist [26]. In the past 11 years there have been many insightful books and many
`detailed articles on process development [27–49].
`Process chemists from AstraZeneca, GlaxoSmithKline, and Pfizer critically assessed
`the parameters for route selection, developing the SELECT acronym: for SAFETY,
`Environmental, Legal, Economics, Control, and Throughput [50]. SAFETY is of
`primary importance. The environmental impact of processing falls under the heading
`of green chemistry. Legal matters are important in the pharmaceutical business, espe-
`cially regarding the issuing of patents and freedom to operate. Economics are expressed
`in the cost of goods (CoG), as influenced by the cost of raw materials, labor, quality
`control (QC), and waste disposal. A primary goal is to minimize the cost of an API to
`$1000–$3000/kg. To minimize the CoG chemistry must be efficient and robust, and
`stringent in-process controls (IPCs) are crucial to ensuring routine operations to produce
`high-quality products. Starting materials and reagents must be subject to competitive
`bidding [51]. Multi-purpose equipment is used in development efforts in order to
`minimize capital investment; when sales of a compound seem more certain purchasing
`dedicated and specialized equipment may be justified. Throughput is reflected in not
`only isolated yields, but also in the amount of product that can be made per unit of
`volume or per unit of time (space–time yield). Manufacturing complexity is minimized;
`for instance, eliminating the use of hazardous or toxic chemicals eases SAFETY
`concerns, and by minimizing processing bottlenecks and the proportion of rejected
`batches that need to be reworked productivity is improved. Throughput and productivity
`are expressions of economics, and these are some of the key drivers for the
`manufacturing of APIs. The SELECT criteria can be considered throughout this book.
`A perspective on process research and development activities is shown in Figure 1.2, in
`which optimization of process development increases going to the right. Based on results
`of screening, a compound may be designated as an NCE and advanced on the pathway to
`becoming an API. As with any general guideline, the development of an API may not
`follow this sketch. For instance, quantities may be prepared in the kilo lab after research
`efforts have identified an optimal route and preferred reagents; in other instances supplies
`of an API suitable to meet the demand for several years may be manufactured in one 5-gm
`batch. The time to progress from an idea to an API may be 5–13 years, or longer.
`Efficient process development begins with drug discovery, and continues through
`routine manufacturing of the drug product [52]. The roles in drug discovery, process
`
`idea
`
`DISCOVERY
`
`batch
`size
`
`mg - g
`
`emphasis:
`
`expedient
`
`PROCESS
`RESEARCH
`KILO
`LAB
`
`PROCESS
`DEVELOPMENT
`
`routine
`e
`g
`manufacturing
`
`kg
`
`1 - 100 kg
`
`> 100 kg
`
`tox batches;
`Phase 1
`
`convenient
`
`Phase 2
`
`Phase 3
`
`practical
`
`efficient
`
`FIGURE 1.2 Perspective on the drug development continuum.
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 7 of 86
`
`

`

`6
`
`Practical Process Research and Development
`
`research and process development are different. Drug discovery chemists design expe-
`dient, diversity-oriented syntheses to prepare many compounds from a common inter-
`mediate. Process research chemists focus on convenient routes to prepare one or perhaps
`a few similar compounds; route selection and reagent selection are usually prime
`considerations. Process development chemists optimize the processes to prepare an NCE
`or API, focusing on minimizing impurities, streamlining work-ups, avoiding chroma-
`tography if possible, and isolating the desired final form of one product [53]. Of course
`these general descriptions may overlap. Almost inevitably process chemists change the
`route developed by the discovery chemist; for a new project one of the initial efforts of the
`process chemist is to consider possible benefits in reordering steps in a route. At some
`point process chemists consider changing the route, using different starting materials or
`taking advantage of biocatalysis. The reactions commonly encountered by researchers
`within pharmaceutical process R&D [54] and drug discovery [55] are relatively similar,
`not surprisingly. The process chemist speeds the development of an NCE by considering
`the “hows” and “whys” of processing. While carrying out the present duties one can
`consider the needs of the next step and thus smooth the development of an NCE. Time
`invested early to make material by processes that can be readily scaled up will shorten
`overall development time.
`Of course the problem with investing time in downstream efforts is that most
`compounds fail to make the marketplace; in 2003, Federsel estimated that less than
`0.03% of all active compounds prepared were approved by regulatory authorities and
`70% of drugs failed to recoup the cost of investments [56]. Perhaps 95% of compounds
`passing through process R&D operations fail. Optimizing processes too early may waste
`resources, but if more material is quickly needed, scale-up may be difficult unless some
`optimization was in place. By Phase 3 the route should be established and process
`optimization should be well underway; if not, supplies for Phase 3 may be at risk.
`Furthermore, the pressure for filing an NDA may result in advancing processes needing
`substantial optimization. Suffice it to say that not all processes are optimized before
`scale-up, and processes may be rushed into production to provide material for clinical
`trials or manufacturing. The conundrum of when to invest in process optimization often
`pivots on the GO/NO GO decision points (Table 1.3).
`“Fit for purpose” has been used and overused to describe efforts to make NCEs. All
`process R&D should be fit for purpose, but usually this phrase is applied to the early
`efforts to make sufficient supplies for toxicology and Phase 1 studies in an expeditious
`fashion. Once proof of safety in humans is established there is usually a need to rede-
`velop processes to make them more efficient and lower the CoG such efforts are also fit
`for purpose [57]. Almost inevitably processes are changed to prepare material for Phase
`3 if not for Phase 2 clinical studies.
`Serendipity should always be exploited. (As Pasteur said, “Chance favors the
`prepared mind.”) For example, the desired dibenzoylated ribonolactone epimer crys-
`tallized and was converted to gemcitabine (Figure 1.3). The benzoyl protecting group
`had been selected for ease of following the reactions by HPLC [58]. Another example
`is the fortuitous epimerization of the methine alpha to the ketone, shown in Figure 1.3.
`After condensation with the ester acid the trans:cis ratio was 35:64, but the ratio
`improved somewhat during acidification for workup. A study showed that the best
`selectivity for the trans-isomer occurred after the basic aqueous phase was held at
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 8 of 86
`
`

`

`Chapter | 1 Introduction
`
`7
`
`TABLE 1.3 Using GO/NO GO Decision Points in Process R&D (1)
`
`GO/NO GO Studies
`
`Amount of API Required Suitable Route
`
`Toxicology studies
`Bioavailability (Phase 1)
`
`Low
`Low
`
`Efficacy (Phase 3)
`
`High
`
`Expedient: consider using
`discovery route and
`preparative chromatography.
`SAFETY concerns may be addressed
`using routine laboratory operations.
`Scale-up can be grueling with only
`minimal experience
`
`SAFETY concerns must be thoroughly
`addressed for scale-up
`Cost-effective synthesis desired
`Controls needed for reliability and filing
`Productive operations desired
`
`(1) Grabowski, E. J. J., Reflections on Process Research II, in Fundamentals of Early Clinical Drug Development;
`Abdel-Magid, A. F.; Caron, S., Eds.; Wiley: Hoboken, NJ; 2006; Chapter 1.
`
`H3C
`
`CH3
`O
`
`O
`
`F
`CO2Et
`
`F
`
`OBz
`
`1) CF3CO2H,
` H2O, CH3CN
`2) Δ, -EtOH
`
`HO
`
`O
`
`BzO
`
`F
`
`1) BzCl
` pyr
` DMAP
`
`2) CH2Cl2,
` heptane
`
`O
`F
`
`4) -40 ºC, H2O
`5) MTBE
`
`organic
`phase
`
`1) (i-Pr)2NH (2.5 eq.)
` THF, -40 ºC
`2) n-hexLi (4.54 eq.)
` - 40 to 3 ºC
`
`3) (1.29 eq.)
`CO2H
` 36 ºC /
` 1.5 hr
`H3CO2C
`
`CH3
`
`HN
`
`O
`
`N
`
`BzO
`
`O
`
`O
`F
`
`BzO
`BzO
`
`+
`
`O
`
`O
`F
`
`F
`BzO
`crystallized
`
`CO2Li
`
`F
`dissolved in
`mother liquor
`
`1) 5 M HCl to
` pH 5.6 - 6.0
`2) hold 0.5 h
`3) 6 M HCl to pH 5.0
`4) H2O wash
`
`CO2H
`
`HN
`
`LiO
`
`N
`
`O
`held at 20 ºC / 8 h
`
`O
`
`HN
`
`O
`
`N
`
`(80%,
`trans:cis 95:5)
`
`FIGURE 1.3 Two examples of serendipity that were exploited.
`
`room temperature for 8 hours [59]. Researchers exploit observations such as these to
`optimize their processes.
`
`II. EQUIPMENT CONSIDERATIONS ON SCALE
`
`Basic operating differences for equipment used on scale are centered on the fact that
`the equipment is opaque and immobile, and there is limited access to the contents. Most
`multi-purpose reactors (Figure 1.4) are made of glass-lined steel, as that material is more
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 9 of 86
`
`

`

`8
`
`Practical Process Research and Development
`
`FIGURE 1.4 Section view of a cylindrical 1000 gallon multi-purpose glass-lined reactor. Heat transfer fluid
`can circulate inside the jacket for cooling/heating capability. Agitator blades are exchangeable. Courtesy of
`De Dietrich Process Systems.
`
`inert than metal. Baffles, vertical blades near the reactor walls, are used to increase the
`turbulence of the agitated mixtures. Once vessels in a pilot plant or manufacturing have
`been charged they are rarely opened; this is to ensure SAFE operations and to prevent
`contamination of the batch. All mixtures must be stirrable for transfer through a bottom
`valve. The agitator is routinely raised above the bottom surface of the reactor, and
`volumes below the agitator cannot be stirred. The minimum agitation volume (Vmin) is
`about 10% of the nominal reactor volume. The maximum volume a vessel can contain
`(Vmax) may be 100–110% of the nominal reactor volume. In a large vessel heat transfer by
`circulating heat transfer fluids through an external jacket is slower, due to the high
`volume and relatively low surface area of the reactor. A concern of using glass-lined
`reactors is that they may fracture due to thermal shock; other reactors may be preferred
`for cryogenic or high-temperature processes. Childers has presented an excellent pictorial
`perspective of large-scale syntheses from the standpoint of a chemical engineer [60].
`Most scale-up costs are associated with mass transfer and heat transfer. Heat
`transfer affects SAFETY and impurities. In semi-batch operations, heat transfer rate
`often determines the time required for operations. Continuous operations may be
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 10 of 86
`
`

`

`Chapter | 1 Introduction
`
`9
`
`are
`Most operations on scale
`possible if time is not an issue, and
`money is not an issue. Rarely are
`there excesses of either. The best
`approach to develop processes that
`can be readily scaled up is to
`mentally scale down operations to
`the laboratory, and mimic them in
`the laboratory.
`
`chosen for fast reactions due to improved
`heat transfer rates on scale. Mass transfer
`often limits the time for operations. Some
`operations cannot be readily speeded up,
`e.g., the time needed to separate immiscible
`phases during extractive work-ups, or the
`time required to transfer
`liquids through
`a 1-inch line. Efficient mixing is needed
`for heterogeneous reactions, such as liquid/
`liquid, gas/liquid, solid/liquid, and multiply
`heterogeneous processes. Efficient mixing
`may be needed for homogeneous reactions
`also; the rate and mode of addition can be crucial, and controlling micromixing and
`mesomixing may be necessary to minimize impurity formation in fast reactions.
`The limitations of heat transfer through external cooling of spherical flasks in the
`laboratory and cylindrical multi-purpose reactors are noticed with the scale-up of batch
`operations. For cylinders and spheres as the radius increases the volume increases faster
`than does the surface area, hence the ability to remove heat by circulating external fluids
`progressively decreases as the volume increases. For a 10-fold scale-up about twice the
`amount of time will be required [61]. Hence rapid additions on scale can lead to uncon-
`trollable exotherms, with degradation of the reaction mixture and possible runaway reac-
`tions. Dose-controlled additions are used to moderate exothermic response from additions.
`On scale additions typically require 20 minutes to hours; pumps are available for extended
`addition times. Longer operations are not necessarily bad for scale-up. Smaller reactors,
`frequently used for continuous operations, provide more rapid heat transfer.
`The scale-up of a Swern oxidation (Figure 1.5) illustrates the extended time required
`for heat transfer and the impact on processing. The benchmarks for the oxidation were the
`laboratory conditions with additions at 15
`
`C. Unfortunately when these temperatures
`were employed in the pilot plant the isolated
`yield was about 60% of the expected yield. In
`the pilot plant about 2 hours was required to add
`both oxalyl chloride and triethylamine, allow-
`ing time for
`the reactive intermediates to
`decompose. By making the additions at 40
`
`C
`the expected yield was achieved [62]. While the
`additions at the colder temperature may not
`have been any faster, the colder temperature
`slowed the decomposition of
`the reactive
`intermediates.
`The Swern oxidation in Figure 1.5 was successfully scaled up later through semi-
`continuous processing, affording essentially the same yield, but with improved process
`
`C, in contrast to the
`control [63] (Figure 1.6). The reaction was carried out at about 40
`colder temperatures used for batch oxidations. The higher reaction temperature was
`made possible by the extremely short residence time (s) of 0.1 seconds for the reaction
`stream in the reactor. (This is an example of the high-temperature short-time (HTST)
`practice for continuous operations, as is used for pasteurizing milk [64].) The reactor
`
`For rapid scale-up, design homoge-
`neous reaction conditions whenever
`possible. Check the stability of
`process
`streams under extended
`conditions in the laboratory before
`scale-up, because processing on
`scale will be extended.
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 11 of 86
`
`

`

`10
`
`Practical Process Research and Development
`
`O
`
`Ph
`
`N
`
`N
`
`O
`
`CO2CH3
`
`O
`
`H+
`
`- H2O
`
`CHO
`
`HN
`
`O
`
`CO2CH3
`
`O
`
`Ph
`
`O
`
`N
`
`OH
`
`1. DMSO, (COCl)2 /
` CH2Cl2
`2. alcohol
`
`3. Et3N / CH2Cl2
`CO2CH3
`
`HN
`
`O
`
`O
`
`Ph
`
`O
`
`N
`
`addition
`temperatures
`
`yield
`
`comments
`
` 2
`
` h each to add (COCl)2 and Et3N allowed
` intermediates to decompose
`faster additions gave higher yield
`
`-15 ºC
`
`-15 ºC
`
`-40 ºC
`
`55%
`
`31%
`
`52%
`
`scale
`
`laboratory
`
`pilot plant
`
`pilot plant
`
`FIGURE 1.5 Scale-up of a Swern oxidation through batch operations.
`
`+ CO + CO2
`
`CH3
`
`H
`
`Cl
`
`OS
`
`H3C
`
`Ph
`
`HO
`
`HN
`
`O
`
`CO2CH3
`
`O
`
`Ph
`
`N
`
`(~220 kg)
`
`transferred into
`Et3N (5.8 eq.)
`at -10 to 10 ºC
`
`HN
`
`O
`
`CO2CH3
`
`O
`
`N
`
`O
`
`CHO
`
`HN
`
`O
`
`CO2CH3
`
`O
`
`O
`
`Ph
`
`N
`
`O
`+ DMSO (10.8 eq.)
` [in CH2Cl2]
`
`O
`
`Cl
`
`Cl
`
`O
` (1.5 eq.)
`+ N2 sweep
`Ph
`
`O
`
`N
`
`N
`
`O
`
`O
`
`CO2CH3
`(61%)
`
`2.1 in ID pipe,
`31.5 in long,
`τ = 0.1 sec
`outlet ~ 40 ºC
`
`1) aq. washes
`2) TFA, Δ
` - H2O azeotrope
`3) Δ, - CH2Cl2
`4) + n-BuOAc
`
`O
`
`NH2
`
`NaOCl
`2 NaOH
`
`H2O
`
`Cl
`NH
`
`O
`
`NaOH
`
`O
`
`C
`
`N
`
`O
`
`H2O
`
`HN
`
`OH
`
`- CO2
`
`NH2
`
`CO2H
`
`O
`
`N
`
`F N
`
`HN
`
`O
`
`HN
`
`NH
`
`ciprofloxacin
`
`Physical separation of process
`streams minimizes urea formation
`
`X
`
`FIGURE 1.6 Large-scale manufacturing by continuous operations.
`
`
`bends in it to increase turbulence for
`was no more sophisticated than a pipe with two 90
`efficient mixing [65]. An example of large-scale manufacturing using continuous
`operations is the preparation of cyclopropylamine, a component of ciprofloxacin and
`some agrichemical products. The Hofmann rearrangement is carried out by continuous
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 12 of 86
`
`

`

`Chapter | 1 Introduction
`
`11
`
`inputs
`
`Batch and semi-batch operations
`
`elapsed
`time
`
`product
`(batch)
`
`product
`(semi-batch)
`
`Inputs
`
`Two continous processing options: CSTR and PFR
`
`continuously
`stirred tank
`reactor
`(CSTR)
`
`plug
`flow
`reactor
`(PFR)
`
`Inputs
`
`Inputs
`
`optional heat
`exchanger
`
`small reactor,
`controlling mixing &
`temperature
`
`Product
`
`Product
`(semi-continuous)
`
`FIGURE 1.7 Batch and semi-batch operations, and continuous operations.
`
`operations with either continuously stirred tank reactors [66] or static mixers [67]
`(Figure 1.7). Chemical engineers in the fine chemicals industry often have a great deal of
`experience with continuous operations.
`The two primary options for operations, batch and semi-batch (or semi-continuous)
`operations, and continuous operations, are shown schematically in Figure 1.7 [68].
`Continuous operations have been used on scale for decades and are slowly being
`incorporated into processes for APIs. Continuous operations are particularly useful
`for fast reactions, those that take place in seconds or minutes as opposed to hours.
`One attractive benefit of these operations is the SAFETY afforded by conducting
`a reaction with only a small amount of a reaction stream at any given moment
`(Chapter 14). Continuous operations are part of process intensification efforts,
`efforts to improve the space–time productivity of processing [69]. Federsel has
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 13 of 86
`
`

`

`12
`
`Practical Process Research and Development
`
`estimated that continuous operations are applied in 10–20% of the processes in the
`fine chemicals and pharmaceutical industry [70]. Roberge and coworkers feel that
`50% of the processes used in the pharmaceutical
`industry could benefit from
`continuous operations [71]. Chris Dowle, of the Center for Process Innovation, has
`stated that “Continuous processing can reduce operating expenses by at least 90%
`and capital expenses by at least 50%” [72]. Continuous operations are not appro-
`priate for every process, but sometimes are the only practical approach to make
`more material.
`
`III. OPERATIONS PREFERRED ON SCALE
`
`Perhaps the first advice that a seasoned industrial process chemist will give to
`someone from academia is that it is not necessary to dry extracts over desiccants such
`as Na2SO4 or MgSO4. This operation may be unnecessary if water is removed as an
`azeotrope during the concentration. As detailed in Table 1.4, a considerable amount
`of time may be expended in drying over desiccants. The cost of this operation can be
`calculated in several ways. The total time required of 16 operator-hours may be
`conservative; this assumes there are no problems, and no supervision is required.
`During the stir-out time of 2 hours other tasks can be undertaken, but the remaining
`steps require some attention. Considering that the cost of plant time may be as high as
`$500–750/h, a substantial amount of money may be spent unnecessarily. But
`
`TABLE 1.4 Estimated Processing Time on Scale for Na2SO4
`Drying Step
`
`Operation
`
`Set up and test filter
`
`Charge Na2SO4
`
`Stir suspension
`
`Filter off solids
`
`Deliver rinse to site
`
`Apply rinse to solids
`
`Suction dry solids
`
`Pack up solids for disposal
`
`Rinse for “SAFE to clean”
`
`Clean equipment
`
`Test equipment for cleanliness
`
`Store equipment
`
`Total
`
`Estimated Operator-hours
`
`2
`
`0.5
`
`2
`
`2
`
`0.5
`
`0.5
`
`0.5
`
`0.5
`
`0.5
`
`4
`
`2
`
`1
`
`16
`
`IPR2020-00770
`United Therapeutics EX2017
`Page 14 of 86
`
`

`

`Chapter | 1 Introduction
`
`13
`
`foremost, as mentioned earlier, this operation may be redundant if a water-solvent
`azeotrope removes water while the extract is being concentrated. The cost of an
`unnecessary step is opportunity cost, for the opportunity lost to accomplish some-
`thing else or learn something else. Hence on scale extracts are rarely dried over
`desiccants such as Na2SO4.
`Table 1.5 compares operations run in the laboratory and on scale. These are
`guidelines suggested for convenient, productive, and cost-effective operations, but most
`of the operations can be run on scale if desired.
`
`TABLE 1.5 Comparison of Process Operations in the Laboratory and on Scale
`
`In the Laboratory
`
`In Stationary
`Equipment, at Least
`50 L Volume
`
`Process
`Operation
`
`Commonly
`Carried Out
`
`Easy
`to do
`
`Commonly
`Carried Out
`
`Easy
`to do
`
`Comments
`
`Drying extracts over
`desiccants, e.g.,
`Na2SO4, MgSO4
`
`Concentrating
`to dryness (1)
`
`Triturating, lixiviating
`(4)
`
`Use of highly
`flammable solvents,
`e.g., Et2O
`
`Decanting
`
`Column
`chromatography
`
`Rapid transfers
`
`Maintain cryogenic
`temperature
`
`Extended additions
`
`Maintain constant
`pH
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X (2)
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X (5)
`
`X
`
`X
`
`X
`
`(3)
`
`X
`
`X
`
`X
`
`X
`
`X
`
`X
`
`Often redundant if
`concentrating removes
`H2O b

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