Вы находитесь на странице: 1из 333

‘No Natural Home’

Placing the Promise of


Biopharming

Richard Milne

Thesis submitted in fulfillment of the requirements for the degree

of Doctor of Philosophy

2009

University College London


I, Richard Milne, confirm that the work presented in this thesis is my own. Where
information has been derived from other sources, I confirm that this has been indicated in
the thesis.
Abstract

Biopharming, or the production of biopharmaceuticals using genetically modified crops,


brings together agricultural and pharmaceutical biotechnologies. It raises hopes of cheaper,
more widely available medicines, and fears of ‗drugs in your cornflakes‘. It prompts new
questions for publics, regulators and researchers and requires reworking and realigning the
arguments, actors and entities which emerged around first generation biotechnologies.

This thesis combines work in science and technology studies and the geographies of
science. It focuses on the role of space and place in establishing and encountering the
promise of biopharming for both publics and experts. The argument draws on research
conducted over three sites: a review of research and regulatory documentation, 13
interviews with researchers involved in an EU sponsored biopharming project and six
repeated focus groups with members of the public in London and North West England.

The thesis argues that the establishment of forceful futures for biopharming is based on the
ability of the technology to successfully take, or make, its place among existing
pharmaceuticals and foods. I describe how the technology‘s promise is performed through
its associations with existing narratives and products. In addition, I suggest that a key step
in establishing and contesting biopharming‘s promise comes through encounters between
its imagined geographies and the places of pharmaceutical and food production. These
elaborate new, contained places of biopharming, neither wholly medical nor agricultural.
The thesis concludes by suggesting that the relationship between the material and imagined
geographies of technological futures warrants further exploration.

3
Acknowledgements

A number of people have contributed a huge amount to the realisation of this thesis. I am
particularly grateful to those who have been central to the research – the biopharming
researchers who kindly gave up their time to be interviewed, and the members of the
public who participated in what were, for me and hopefully for them, stimulating and
rewarding focus group meetings.

I am indebted to my supervisors Dr Brian Balmer and Dr Gail Davies. I cannot thank them
enough for the endless and enthusiastic encouragement and support they have provided
throughout the planning, execution and writing up of my research. Their comments and
suggestions have helped me greatly in the development of my thesis.

I also wish to thank the staff and research students of the UCL Institute of Human Genetics
and Health. Their diverse perspectives and wealth of knowledge in the social and natural
sciences have been of huge benefit to me in exploring the potential, boundaries and
limitations of my research.

I am grateful to the staff and students of the Science and Technology Studies and
Geography departments at UCL, not least for the regular opportunity to present, discuss and
develop my work. I would also like to acknowledge those who provided valuable
comments at the meetings of 4S, the RGS/IBG and the BSA, as well as at Cesagen in
Lancaster.

Finally, I wish to thank those close to me who have helped and encouraged me, and without
whom the process would have been infinitely more arduous, not least my parents and my
sister Heather. Most of all, I would like to thank Sophie whose love, patience and joie de
vivre have made my PhD so enjoyable, and whose questioning, discussion and proof
reading have improved it enormously.

4
Table of Contents

List of Figures ...................................................................................................................... 10

List of Abbreviations............................................................................................................ 11

CHAPTER 1: NO NATURAL HOME? .......................................................................... 12

1.1 Introduction ............................................................................................................... 12

1.2 From Cohen and Boyer to Crops and Biologics: the biotechnological context ........ 18

1.3 From Biologics to Biopharmaceuticals ..................................................................... 23

1.3.1 Challenging Pharmaceutical Promise ................................................................ 26

1.4 Agricultural Biotechnologies: Plants and Publics ..................................................... 30

1.5 Biotechnologies' Publics ........................................................................................... 36

1.6 Biopharming.............................................................................................................. 40

1.6.1 The Prodigene Fiasco .............................................................................................. 45

1.6.2 The Pharma-Planta Consortium .............................................................................. 47

1.7 Relating Science and Society .................................................................................... 54

1.8 Places and Placing ..................................................................................................... 59

1.9 Thesis Outline ........................................................................................................... 61

CHAPTER 2: OF PLACING AND PROMISING .......................................................... 64

2.1 From Efficacy to Affectivity .......................................................................................... 65

2.1.1 The Imagination and Technological Futures ........................................................... 66

2.1.2 Promising Communities .......................................................................................... 69

2.1.3 Promising Geographies ........................................................................................... 72

2.1.4 From Imagined Futures to Authorising Pasts .......................................................... 74

2.2 Making Expectations Matter .......................................................................................... 78

2.2.1 The proof of the pudding is … in structural and functional similarity? .................. 81
2.2.2 The proof of the pudding is … in looking and touching? ....................................... 83

2.2.3 The importance of association and the problems of consumption .......................... 86

2.2.4 Between Consumers and Patients? .......................................................................... 88

2.3 A Place for Everything, and Everything in its Place ...................................................... 92

2.3.1 Places of Technoscientific Quality .......................................................................... 95

2.3.1.1 The Contested Spaces of Agricultural Biotechnology ...................................... 99

2.3.1.2 The Emerging Geographies of Pharmaceutical Biotechnology ...................... 101

2.3.2 Quality and Agricultural Place .............................................................................. 103

2.3.3 Combining the Spaces of Food and Pharmaceuticals............................................ 105

2.4 Summary and Conclusions ........................................................................................... 106

CHAPTER 3: QUALITATIVE METHODS AND PLACING BIOPHARMING ..... 108

3.1 Focussing on Groups .................................................................................................... 111

3.1.1 Introducing Group Methodologies ........................................................................ 111

3.1.2 Who to Recruit? Naturalism and Pragmatism ....................................................... 115

3.1.3 Recruiting the Research Groups: Heterogeneity, Homogeneity and Hard Work . 117

3.1.4 Practice of Groups ................................................................................................. 122

3.2 Interviews and research documents ............................................................................. 128

3.2.1 Selecting Interviewees ........................................................................................... 130

3.2.2 Recruiting PPC Members ...................................................................................... 132

3.2.3 Just Talk? Research Documents and Reviews as Narrative .................................. 137

3.3 Engagement between Experts and Publics: ELSI and the Media ................................ 139

3.4 Analysing The Data...................................................................................................... 142

CHAPTER 4: FAMILIAR NARRATIONS AND PLACED EXPECTATIONS ....... 146

Part I Familiar Narrations................................................................................................... 147

4.1 Experts: From Agriculture to Biopharmaceuticals ...................................................... 147


6
4.1.1 Narrative One: From Agriculture to Food ............................................................. 147

4.1.2 Narrative Two: From Biotechnology to Botany.................................................... 151

4.2 Publics: The Serendipitous Pasts of Biomedicine ........................................................ 158

Part II Placed Expectations ................................................................................................ 165

4.3 Experts: Philosophy and Profit? ................................................................................... 165

4.3.1 Imagined Geographies and Technical Solutions ................................................... 166

4.3.2 Plant Made Profits? ............................................................................................... 171

4.4 Publics: Pharmaceutical Promises................................................................................ 173

4.4.1 Technical Promise and Social Failure? ................................................................. 174

4.4.2 Profits and the Purpose of Pharmaceutical Production ......................................... 181

4.5 Summary and Conclusions ........................................................................................... 188

CHAPTER 5: BIOPHARMING MATTERS ................................................................ 191

5.1 Experts: The Promise of Safe Products ........................................................................ 192

5.2 Consuming Foods ........................................................................................................ 196

5.3 Consuming Pharmaceuticals ........................................................................................ 204

5.3.1 Natural Medicines ................................................................................................. 209

5.4 Experts: The Problem of Pharmaceutical Equivalence ................................................ 214

5.5 Publics: Troubled Consumption................................................................................... 220

5.6 Re-establishing Distinctions between Pharmaceuticals from Food Products? ............ 225

5.7 Summary and Conclusions ........................................................................................... 230

CHAPTER 6: PLACING PHARMACEUTICAL PRODUCTION ............................ 233

6.1 The Landscape of Foods and Pharmaceuticals in Public ............................................. 234

6.1.1 The Places of Food ................................................................................................ 234

6.1.2 Pharmaceutical Places ........................................................................................... 237

6.2 Biopharming‘s Public and Expert Places ..................................................................... 240


7
6.2.1 Publics: The Pharmaceutical Places of Biopharming ............................................ 240

6.2.2 Publics: Biopharming and Food Spaces ................................................................ 243

6.2.3 Experts: Biopharming and Agricultural Space ...................................................... 246

6.2.4 Experts: Spaces of Pharmaceutical Concern ......................................................... 250

6.3 Reconciling Agricultural and Pharmaceutical Spaces ................................................. 257

6.3.1 Publics: Located Promise ...................................................................................... 257

6.3.2 Experts: Greenhouses and GMP............................................................................ 263

6.4 Resolving the Places of Biopharming, ......................................................................... 269

6.5 Summary and Conclusions ........................................................................................... 273

CHAPTER 7: NO PLACE LIKE HOME...................................................................... 276

7.1 Summary and Synthesis ............................................................................................... 277

7.2 Exploring the Geographies of Biopharming ................................................................ 281

7.2.1 Imagined Geographies of the Future ..................................................................... 282

7.2.2 Socio-Spatial Geographies of Expecting ............................................................... 284

7.2.3 Obdurate Geographies ........................................................................................... 286

7.3 Looking Beyond the Thesis ......................................................................................... 288

7.4 Future Directions .......................................................................................................... 290

7.5 Placing the Promise of Biopharming ........................................................................... 291

APPENDIX I...................................................................................................................... 293

APPENDIX II .................................................................................................................... 300

APPENDIX III ................................................................................................................... 302

APPENDIX IV ................................................................................................................... 307

BIBLIOGRAPHY .............................................................................................................. 310

8
9
List of Figures

1.1 The intersection of biopharming with biotechnology


1.2 A timeline of significant events in the development of red and green
biotechnologies
1.3 European Regulations and Directives relevant to Pharmaceutical Biotechnology
1.4 European Regulations and Directives relevant to Agricultural Biotechnology
1.5 Milestones in Biopharming
1.6 The aims of the Pharma-Planta Consortium
1.7 Three Models of the Science and Society Relationship

2.1 Simplified knowledge topologies and black-boxes for different types of food
consumption

3.1 Focus group constitution


3.2 The Ginster‘s Cornish Pasty
3.3 The structure of the Pharma-Planta Consortium
3.4 The distribution of PPC institutions
3.5 Distribution of researchers interviewed
3.6 Researchers interviewed

4.1 Summary of Chapter Four

5.1 Pills of crushed plant leaves


5.2 St John's Wort in tablet form
5.3 Antibody producing maize kernels tagged with DsRed viewed under normal and
UV light.
5.4 Summary of Chapter Five

6.1 The GMP Facility, Fraunhofer IME, Aachen


6.2 Greenhouses at Fraunhofer IME, Aachen
6.3 The pressurised corridor leading to research chambers in the Unigro Grodome
6.4 Summary of Chapter Six

10
List of Abbreviations

CHO Chinese Hamster Ovary

DEFRA UK Department for the Environment, Food and Rural Affairs

EFSA European Food Safety Agency

EMEA European Medicines Evaluation Agency

GACP Good Agricultural and Collection Practice

GM Genetically modified

GMP Good Manufacturing Practice

HSE UK Health and Safety Executive

NGO Non-Governmental Organisation

PMP Plant Made Pharmaceutical

PPC Pharma-Planta Consortium

11
CHAPTER 1: NO NATURAL HOME?

1.1 Introduction

In July 2004, the European Union‘s Sixth Framework Programme awarded 12 million
euros to a European partnership for the development of a plant made pharmaceutical
product to the clinical trial stage. The press release accompanying the launch of the
‗Pharma-Planta Consortium‘ (PPC) described:

―a major new research program to explore the use of genetically modified plants in
the development of treatments for some of mankind's most devastating diseases,
including AIDS, diabetes, rabies and tuberculosis‖(PPC 2004)

The consortium initially targeted the production of a microbicidal HIV cream in maize
(Ramessar, Rademacher, et al. 2008), and post-bite treatments for rabies in tobacco. In its
use of genetically modified crops, biopharming or ‗molecular farming‘ is an extension of
agricultural (green) biotechnology. However it is also a biomedical (red) application of
biotechnology, targeting the development of novel pharmaceuticals.

The PPC press release echoes the 1970s ‗genetic engineering gold rush‘ (Wright 1994) in
promising advances in medicine and agriculture. Such promises are an inextricable part of
new biotechnologies. As anthropologist Sarah Franklin describes, ―the importance of
imagining a future yet-to-be … fundamentally defines the whole issue of the new genetics
and society‖ (Franklin 2001:349). Technological promises and expectations mobilise both
public and investor support, and shape technical choices and regulatory and institutional re-
orderings. In this thesis, I explore the promise of ‗biopharming‘, examine how these
imagined futures shape the technology‘s development, and demonstrate how they are
challenged and transformed as they engage with and change the narratives, materials and
places of existing pharmaceuticals and foods.

Since the 1970s, agricultural and pharmaceutical biotechnologies have taken different
paths. They have been incorporated into existing social, technical and institutional networks

12
of their fields of application, and while reshaping these, have themselves been transformed.
Efforts to bring them together have met with little success. In the 1990s, corporate visions
of a unified biotechnological future for the ‗Life Sciences‘ foundered. A division between
red and green represents the ‗new strategic context‘ (Bauer 2002) for biotechnology, as
illustrated by their contrasting fortunes in the UK.

Less than six months prior to the Pharma-Planta announcement, in March 2004, Bayer
Cropscience decided to abandon its application to grow genetically modified (GM) maize
in the UK. In June this was followed by Monsanto‘s decision to discard plans to introduce
GM wheat, not just in the UK but worldwide. In an article for the UK‘s Guardian
newspaper, Sue Mayer, director of NGO Genewatch UK asked ‗Is this the end for GM
food?‘ (Mayer 2004).

The ‗end of GM‘ described by Mayer was the culmination of nearly a decade of
controversy. In the late 1990s, stabilised regulatory regimes around biotechnology, genetics
and food in the UK and across Europe had been thrown into turmoil in the face of public
opposition. Mayer suggests that ―the scale and trajectory of GM crops and foods has been
changed‖ through the achievements of ―ordinary people and campaigners‖ (Mayer 2004).
Public concerns around GMOs concentrated not only on scientifically defined risks, but on
the institutional contexts, motives and justifications of their development (Mayer and
Stirling 2004). As Sheila Jasanoff describes in her study Designs on Nature, the evolution
of the GM controversy in the UK led to reworking of relationships between science and
society (Jasanoff 2005).

However, other applications of genetic science have been less critically received in the UK.
In August 2004, the UK became the first European country to grant a licence for
therapeutic human cloning for stem cell research. The chair of the Human Fertilisation and
Embryology Authority, Suzi Leather, described how this was ―an important area of
research and a responsible use of technology" and emphasised that the licence had been
granted after consideration of ―all the scientific, ethical, legal and medical aspects of the
project‖ (in Bhattacharya 2004).

13
The juxtaposition of these events, just months apart, show the persistently contrasting
fortunes of medical (red) and agricultural (green) biotechnologies in the UK since the
‗watershed years‘ of the cloning of Dolly and imports of GM soya in the late 1990s. In the
seven years after Dolly the sheep, the UK polity arrived at a point at which the cloning of
human embryos could be justified on a biomedical basis. Yet, after a decade, attempts to
introduce biotechnological foods continued to struggle.

Together, these events bracket the launch of the PPC, and effectively demonstrate the
complex environment for new biotechnologies. Moreover, they highlight the insufficiency
of technologically deterministic accounts of technological advance. They emphasise that
the logic of biotechnology‘s development is not inherent to it, and instead is produced
together with reconfigurations of existing social, political, economic and ethical
relationships. Researchers in science and technology studies have described this
relationship between science and society and between public and expert knowledges as one
of ‗co-production‘ (Jasanoff 2004) or ‗co-construction‘ (Irwin and Michael 2003).

A comparison between the UK and the US further illustrates this ‗co-production‘ of the
scientific and the social and its geographical distribution. As Jasanoff (2005) describes, the
British experience with both GM and stem cell research contrasts markedly with that of the
United States. In January 2001, George W. Bush had devoted his first televised address to
the withdrawal of federal funds from stem cell research. Yet in 2004, while Bayer were
withdrawing from the UK, the USA planted 47.6 million hectares of genetically modified
crops, 59% of the global GM area (James 2004). The comparison emphasises that both the
rejection of GM crops in Britain and the relative acceptance of therapeutic cloning for stem
cell research are an outcome of British and US ‗political cultures‘ (Jasanoff 2005), rather
than inherent to the technologies themselves.

A short EMBO Report published by members of the PPC to accompany its launch
recognised the heterogeneous challenges of being both ‗red‘ and ‗green‘ in the UK and in
Europe (Ma, Barros, et al. 2005). It suggested that the realisation of biopharming‘s
potential would come not only through technical success, but through public acceptance
and the development of new regulatory orders.

14
PPC researchers describe how biopharming has ‗no natural home‘ in an EU regulatory
system divided between red and green applications (Ma, Barros, et al. 2005; Sparrow et al.
2007). They suggest the need for regulatory innovation to accompany the scientific work of
the PPC. However, debates around first generation genetically modified crops have
demonstrated that achieving regulatory success alone is insufficient for the successful
introduction of new technologies. A ‗second hurdle‘ has been added, that of public opinion
(Bauer and Gaskell 2002), as recognised in the conclusions of the EMBO report:

―The media-led furore over GM food in Europe illustrated very clearly that although
research and development success and social acceptance are both necessary for the
success of new biotechnology, social rejection alone is sufficient to derail the
endeavour‖
(Ma, Barros, et al. 2005:597) .

The recognition of the importance of both technical and social factors in the development
of new technologies is clearly a result of controversies around the introduction of
genetically modified foods. The relationship of biopharming with agricultural
biotechnology makes public perception an obstruction to realising the pharmaceutical
potential of the technology. However, at the same time, Ma and colleagues argue, the
pharmaceutical nature of the technology has the potential to change perceptions of
agricultural biotechnology. Biopharming:

―offer[s] a real opportunity for the public to see a distinct, advantageous end use for
this technology, with the ability to produce therapeutic drugs more efficiently and
with less expense‖
(Ma, Barros, et al. 2005:598)

Biopharming represents both the combination and extension of agricultural and


pharmaceutical applications of biotechnology. In Europe its development is taking place
among regulations which separate red and green applications, and publics sceptical about
agricultural biotechnologies, but positive about biomedical applications (Gaskell et al.
2006). Bringing these together involves challenges as biopharming engages with a diverse
range of existing social, technical, economic and political processes. It involves the

15
development of new interconnections and interrelations between agricultural and
pharmaceutical actors and entities.

Hence, biopharming represents the intersection of multiple socio-technical regimes. In


work exploring US public attitudes to plant made vaccines, Kirk and McIntosh (2005)
present this intersection graphically. In this depiction, reproduced in Figure 1.1 below,
biopharming is placed at the intersection of agriculture, pharmaceuticals and biotechnology.
It incorporates aspects of agricultural biotechnology, biopharmaceutical manufacture and
medicinal plants, yet is distinct from each. In the discussion that follows I introduce these
medical and agricultural contexts before turning to biopharming itself.

Figure 1.1 The intersection of biopharming with biotechnology (from Kirk and
Mcintosh 2005)

However, this diagram is not without its flaws. It artificially bounds discussion of
biopharming. It concentrates only on the technologies of each field, while agricultural and
pharmaceutical applications of biotechnology have been characterised by the ‗co-
production‘ (Jasanoff 2004) of the social and the technical. Moreover, it occludes the
relationship of both biopharming with the practices and institutions of everyday food and
pharmaceutical consumption and production that I explore in my research.

16
Using the PPC as a case study, this thesis draws on the sociology of science and technology
and the geographies of science to explore how the meanings and futures of biopharming
perform, and are performed through, the narratives, materials and in particular the
geographies of food and pharmaceutical.

I focus on two empirical sites, bringing together expert and public perspectives on
biopharming. The first site is the research of the consortium itself, in the form of analysis of
research and review papers, and interviews with biopharming researchers. These focus
particularly on the production of the HIV microbicide in maize, which forms the ‗fast track‘
of the PPC‘s research programme (PPC 2007). I examine how the materials, narratives and
places of the new technology mobilise and embody imagined futures and geographies of
developed and developing world medicine.

The second site is a series of repeated focus groups with members of the public. The first of
these considers public discussion around conventional foods and pharmaceuticals, along
with previous biotechnologies. The second meeting discusses biopharming, and allows the
new technology‘s relationship to, and reworking of, everyday foods and medicines to be
explored.

In the following sections I introduce the ‗biotechnological context‘ of the technology in


both pharmaceutical and agricultural applications. In section 1.5 I describe the relationship
between the publics of red and green biotechnologies, before introducing biopharming
itself. This provides a basis for the discussions of biopharming which are expanded in the
empirical chapters of the thesis. I then introduce my approach to the relationship between
science and its publics, expanding discussion of co-production and co-construction. Finally,
I consider the usefulness of ‗placing‘ as a heuristic for exploring the relationship between
biopharming and red and green biotechnologies and outline the approach of the thesis.

17
1.2 From Cohen and Boyer to Crops and Biologics: the
biotechnological context

Historian of biotechnology Robert Bud suggests that two dominant understandings of


biotechnology exist, one seeing it as ―just invented‖, another as ―derived from the
Babylonians‖ (1991: 416). For example, the history of biotechnology provided by industry
body BIO (2008) dates back 10,000 years, to the first cultivation of potatoes and the
domestication of livestock. The timeline they present becomes increasingly crowded as it
moves through the 19th and 20th centuries. Robbins-Roth‘s (2001) timeline of
biotechnology, presented as an appendix to her book ―From Alchemy to IPO‖, starts with
the foundational genetic work of Gregor Mendel in 1866, but then skips rapidly to the
1940s and 1950s and the elaboration of the structure and function of DNA. An even shorter
narrative is provided by a forward-looking 2003 industry piece, which commemorates only
the 20th anniversary of biotechnology and looks forward to the next 20 years (Nagle et al.
2003).

The importance of narratives in the governance of the new biotechnologies is described in


detail in Sheila Jasanoff‘s (2005) discussion of how biotechnology was ‗framed‘ in
different ways in 1980‘s Germany, the UK and the US. These framings inserted
biotechnology into existing storylines and differentiated it from others. Regulators thus
positioned the novel ontologies of biotechnology ―either on the side of the familiar and
manageable or on the side of the unknown and insupportably risky‖ (Jasanoff 2005:139).

In the UK, biotechnology was initially differentiated from previous production processes.
Early policy framings emerged from the report of the Royal Commission on Environmental
Pollution, which placed the potential risks of agricultural biotechnology within the context
of previous experience with invasive plant species. Together with wider EU regulation, this
approach brought ecologically oriented expert perspectives together with a ―normative
posture which admitted more uncertainties and called for a precautionary approach to
regulation‖ (2005: 275). As Jasanoff puts it, following early UK reviews and reports on
agricultural biotechnology ―the theme of nature‘s unpredictability echoed through official
British policy‖ (2005: 57).
18
In Germany, governance responses were more prohibitive, as biotechnology was received
with a suspicion derived from the nation‘s past experiences of the abuse of genetic
technologies and the presence of a strong environmental representation in government.
Biotechnology was presented by NGOs and Green politicians as part of a programme of
human interference in nature and as a dangerous alliance between science and the state.
This prompted large scale discussion of risks, broadening the scope of those engaged
beyond traditional experts to focus on the value implications of biotechnology (Jasanoff
1995). In response, regulatory agencies attempted to instate ‗categorical bright lines‘ and
prevent ‗conceptually messy situations‘ (Jasanoff 2005).

In contrast, in the United States, regulators fitted biotechnology into existing regulatory
structures, arguing that there was no reason to distinguish between it and conventional food
and pharmaceutical manufacturing processes. As Plein (1991) observed, lengthy histories
of biotechnology were mobilised in early regulatory debates to establish familiarity and to
normalise the new technology, reducing the regulatory burden. The history of exotic and
invasive species deemed relevant in the UK was ruled out. For US regulators biotechnology
was framed as ―a supplier of familiar classes of products – not as a novel technological
process‖ (Jasanoff 1995:316). Genetic modification was ‗black-boxed‘ (Latour 1988) as
precise, predictable and as indistinguishable from conventional farming.

However, appearing too continuous with existing technologies and products presents
problems in claiming novelty and consequently in establishing intellectual property rights
and garnering financial and public support. As the Biotechnology Industry Organization
asks after introducing its genealogy of biotechnology:

―Crops? Cheese? That doesn‘t sound very exciting. So why does biotechnology
receive so much attention?‖(BIO 2008:1)

In answering this question, BIO suggests the last 40 years have seen a shift of scale, from
‗macro level‘ biotechnology to a ‗micro level‘. While recognising the difficulty, if not
impossibility, of establishing a satisfactory narrative of biotechnology, this thesis takes as
its starting point this shift in scale through biotechnology‘s ‗wedding with genetics‘ (Bud
1991) in the 1970s, and the increase in ‗excitement‘ it prompted.

19
The distinctive ways in which initial developments in biotechnology were received, and the
emergence of divisions between agricultural and pharmaceutical biotechnologies,
emphasises the intersection between biotechnology and social, economic and political
orders. Product, process and programme-based framings allowed the ‗normalisation‘ of
biotechnology within each nation, and provided the basis for the classification and control
of biotechnology and its risks.

Bearing in mind the performative and framing consequences of narrating biotechnology, in


figure 1.2 I present a partial timeline of biotechnology since the development of
recombinant DNA techniques in 1973. The narrative presented by my timeline is one of the
‗breakthroughs‘ through which biotechnology has leapt forward. Such timelines of
biotechnology have become a literary form of their own, with a central role in the narration
and self-presentation of research (Hilgartner 2006). They present an unavoidable sense of
progress and an image of inexorable and entwined technological and economic advance as
material power is transformed into economic power (Wright 1994). They perform a lineage
of development that provides the basis for the extrapolation of hopeful futures, and which
writes out the failures of past futures. However, even within these linear depictions, it is
impossible to escape the co-production of technical successes of biotechnology with
emerging regulatory, economic and social orders.

As does a similar timeline presented by BIO (2008), my timeline presents significant events
in agricultural (‗green‘) and biomedical (‗red‘) biotechnologies alongside one another. Yet
BIO themselves suggest that rather than talk about ‗biotechnology‘ it may be better to
discuss ‗biotechnologies‘ (BIO 2008). My timeline stops at the 2004 events described
above. From here, the discussion of my thesis takes over, focussing on the development of
biopharming from 2004 until summer 2008.

20
Figure 1.2 A timeline of significant events in the development of red and
green biotechnologies

1973 Cohen and Boyer produce the first recombinant DNA molecules. ―This
invention…is the basis for the entire biotech revolution‖ (Robbins-
Roth 2001:220)
1974 The development of hybridoma technology and the production of
monoclonal antibodies.
1975 The ‗Berg Letter‘ prompts self-regulation of the scientific community
at Asilomar and a moratorium
1978 Genentech expresses human insulin in E.coli bacteria
1982 Genetically engineered insulin (Humulin) produced by Genentech and
Eli Lilly approved by US FDA
1983 First genetically modified plant (petunia) produced
1985 Recombinant Human Growth Hormone approved by FDA
1986 The first monoclonal antibody treatment.
The first field tests of genetically engineered tobacco plants
1987 Field trials of ‗Ice-Minus‘ (or ‗Frostban‘) bacteria to prevent frost
damage to strawberries prompt controversy
1989 Field tests of first genetically modified food crop – Calgene‘s GM
tomatoes
1990 First field trials of genetically modified, herbicide tolerant maize
1993 OECD outlines the basis for ‗substantial equivalence‘ - similarity to
existing products establishes the ‗relative‘ safety of genetically
modified foods
1994 Regulatory approval of the first genetically modified food, Calgene‘s
‗FlavrSavr‘ tomato
1996 First commercialisation of genetically modified crops,
Introduction in the UK of GM tomato puree
Monsanto‘s GM soya, mixed with conventional soya, is met with
protests by Greenpeace UK
1997 Cloning of Dolly
1999 Monsanto abandons plans to develop Genetic Use
Restriction/‗Terminator‘ Technologies
Losey et al. (1999) suggest that GM pest-resistant maize pollen is toxic
to Monarch butterfly larvae
2000 ‗Golden Rice‘ engineered to combat vitamin A deficiency
2001 Quist and Chapela suggest that DNA from GM maize has moved into
indigenous Mexican maize
2003 UK ‗GM Nation?‘ consultation
2004 UK authorisation granted for Bayer‘s Chardon LL maize. Bayer
subsequently withdraws its application

21
The early ‗genetic engineering gold rush‘ (Wright 1994) concentrated on pharmaceutical
and agricultural applications. Their development was accompanied by hopeful and fearful
visions of the futures the new technologies would bring. Both forms of future have been
and continue to be characterised by (and caricatured as) ‗hype‘ (for example by Burke
2004; Robinson 2008). The suggestion is that these visions are mere rhetoric, unsupported
by the actual facts of biotechnology. However, these attempts to ‗colonise‘ the imagined
futures of new technologies have important material consequences for their development
(Brown and Michael 2003; Hedgecoe and Martin 2003).

Biotechnology‘s initial introduction to agriculture was accompanied with significant


fanfare. Kloppenburg (2004) cites Kenneth Frey‘s hyperbolic 1984 presidential address to
the American Society of Agronomy as a prime example:

―Let your imagination roam…The future for agronomy is not only bright, but it has
no foreseeable bounds‖ (Frey 1985 in Kloppenburg 2004: 193)

For agriculture, recombinant DNA technologies offered the promise of outdoing evolution.
Researchers foresaw a ‗Doubly Green Revolution‘ (Conway 1999), whereby biological and
geographical restrictions on crop production would be loosened or removed so that ―in 5 to
10 years, Saudi Arabia may look like the wheat fields of Kansas‖ (Mintz 1984 in
Kloppenburg 2004: 203).

Descriptions of the potential of biotechnology in agriculture were paralleled, and indeed


exceeded, by the promise of a ‗biotechnological revolution‘ in medicine. These visions
incorporated the elucidation and targeting of genetic causes of disease, and the manufacture
of new medicines. For Walter Gilbert, the Nobel laureate president of Biogen, the
biotechnology revolution involved the:

―the promise of a new kind of medicine that is based on the natural human proteins;
a medicine that fights disease not by the small molecules made by chemistry, but by
the natural body compounds made by the most modern technology‖ (in Bud
1991:441).

For Gilbert, biopharmaceuticals were the coming together of pharmaceutical science and
‗natural‘ medicine. They represented the exploitation of the compounds of the body
22
through their separation from these bodies, and a renewal of the early 20th century synthetic
chemical revolution in pharmaceutical manufacture.

In the following sections I explore the development of agricultural and pharmaceutical


biotechnology as these hopeful futures established their place among the products, publics
and places of agriculture and medicine. This provides the background for my introduction
of biopharming.

1.3 From Biologics to Biopharmaceuticals

Over the last century, there has been a move from ‗biologics‘, medicines extracted from
blood or other biological sources, to ‗biopharmaceuticals‘, a term coined in the 1980s to
describe the products of recombinant DNA and hybridoma technologies. Pisano (2002)
suggests that the introduction of biotechnology to pharmaceutical research and
development cannot be seen as a single event, but is three interrelated processes. These
include the use of the new genetics to search for novel therapeutic targets, and the
stratification of both patients and diseases through pharmacogenetics (Hedgecoe and Martin
2003; Smart and Martin 2006). They also include the ‗high throughput screening‘ of new
therapeutic compounds for pharmaceutical activity prior to manufacture. However, in this
thesis I focus on the role of biotechnology in the synthesis of therapeutic compounds. As
this has been discussed in less depth by researchers in science and technology studies, I
spend some time introducing it here.

The early development of the pharmaceutical industry relied on the extraction of


pharmaceutical compounds from plants. However, Walsh (2007) suggests that only four
pharmaceuticals that targeted specific diseases were available at the turn of the 20th
century: digitalis, for heart disease; quinine, for malaria; pecacuanha, for dysentery; and
mercury, for syphilis. Of these, three are plant derived.

These early medicines drew on specific geographies, natures and political and economic
networks. The long tradition of ‗physic‘ gardens, including those in monasteries, was
driven by need to provide medicinal products (Livingstone 2003). Later, centres such as

23
Kew Gardens provided conduits for the passage of pharmaceutical plants such as cinchona
for quinine from South America to India, enabling the expansion and consolidation of
British rule (Withers 2002).

In the early 20th century, synthetic chemistry emerged as a means of improving on these
‗natural‘ remedies, and separating them from their places of production. From 1899, aspirin
no longer needed to be extracted from willow bark, but could be synthesised by the dye
manufacturing company Bayer. In the process, chemical synthesis established the pill as a
convenient way to administer standardised doses of chemical drugs.

Alongside this work, anti-toxin sera such as that for diptheria introduced a ‗biological
heuristic‘ (Hopkins et al. 2007) based on the exploitation of complex biological molecules.
Combining these extractive and biological models provided a stream of new therapies
derived from animals, notably insulin, but also hormones and steroids, some of which
began to be produced synthetically by the 1930s (Hopkins et al. 2007).

The 1930s saw the expansion of the pharmaceutical industry, building on the discovery of
drugs such as penicillin and the development of organic chemistry based ‗sulfa‘ drugs,
derived from the red dye prontosil rubrum (Walsh 2007). The development of the ‗chemo-
therapeutic‘ revolution following World War II involved significant investments in research
capacities in organic chemistry and later virology, biochemistry and enzymology
(Galambos and Sturchio 1998; Pisano 2002).

It was against this background that the new biotechnology and biopharmaceuticals emerged
in the 1970s. In Walsh‘s (2007) introduction to pharmaceutical biotechnology, the impact
of the new biotechnology is described as four-fold:

1. It overcomes the problem of source availability.


2. It overcomes problems of product safety.
3. It provides an alternative to direct extraction from inappropriate/dangerous source
material.1

1
Walsh gives the example of the anticoagulant protein Ancrod, produced in the venom of the Malaysian pit
viper. As Walsh puts it: ―although retrieval by milking snake venom is possible, and indeed may be quite an
exciting procedure, recombinant production…would be considered preferable by most‖ (Walsh 2007:5)
24
4. It allows the engineering of therapeutic proteins displaying advantages over the
native protein product.

Biotechnology‘s promise is based on the ability to produce existing biological medicines on


a large scale, safely and more easily. It also offers the potential to develop new
pharmaceutical proteins, and improve on nature. For example, the modification of amino
acid sequences can be used to produce faster acting insulin. The introduction of rDNA and
hybridoma technologies for antibody production thus revived the biological heuristic of
pharmaceutical development (Hopkins et al. 2007) and provided the basis for the
development of a pharmaceutical biotechnology industry.

The development of monoclonal antibodies (MAbs) extends and refines earlier anti-sera
techniques. Antibodies, which bind specifically to antigens on the surface of cells, allow
highly accurate diagnostic tests and the specific targeting of drug treatments. Those derived
from animal sources are ‗polyclonal‘ - they are derived from multiple types of cell - and are
difficult to extract in large quantities. The development of ‗hybridoma‘ technology by
Kohler and Milstein in 1975 allowed the production of antibodies in large numbers.
Hybridomas are ‗immortal cell lines‘ formed from the fusion of tumour cells with
lymphocytes. They can produce large quantities of MAbs derived from a single cell. MAbs
are the largest category of biopharmaceuticals under development (Walsh 2007), and
Hopkins et al. conclude that they ―are the best indication yet of biotechnology‘s long-term
promise in the development of new products‖ (2007: 577).

Recombinant DNA techniques provide new tools for pharmaceutical research and the
manufacturing of pharmaceutical proteins previously extracted from animals. The
movement of genes between bacteria in 1973, and the expression of Xenopus toad genes in
E.coli in 1974, laid the groundwork for the bacterial synthesis of proteins (Wright 1994).
The first human medicine to be produced was insulin. Although the amino acid structure of
insulin is almost conserved across species, allowing the traditional use of pig or cow
insulin, a combination of biological, geographical and socio-cultural factors contribute to

25
favouring recombinant production, as Walsh (2003) describes in an earlier introduction to
pharmaceutical biotechnology:

"The quantity of purified insulin obtained from the pancreas of one pig satisfies the
requirements of one diabetic for 3 days. The supply of pancreatic tissue is dependent
upon the demand for meat, which does not necessarily correlate with the increasing
worldwide incidence of diabetes. Recombinant DNA technology provides an obvious
way to ensure future adequate supply of insulin" (Walsh 2003: 313)

The availability of porcine insulin is limited both by the natures of pigs themselves, and the
geographical and cultural distribution of food. The early promise of recombinant
production saw it as breaking free from these restraints. Insulin was synthesised by
Genentech in 1977 (Wright 1994). As historian Robert Bud describes, the implications of
this synthesis were seen ―not merely as launching a new product, but also as beginning a
new world‖ (Bud 1993:181).

In 1982 Genentech‘s ‗Humulin‘, produced in collaboration with Eli Lilly, became the first
biopharmaceutical to be approved by regulatory agencies. Since then, a number of
recombinant proteins have been approved, the vast majority produced in E. coli bacteria,
the yeast S. cerevisiae or in animal cell lines, predominantly Chinese hamster ovary (CHO)
cells or baby hamster kidney cells (Walsh 2007). However, as I describe in the following
section, the introduction of biopharmaceuticals has not been as smooth as first imagined.

1.3.1 Challenging Pharmaceutical Promise

Along with human growth hormone and erythropoietin (a hormone involved in red blood
cell formation), insulin formed the first stream of biotech products. However, by 2006 only
160 biopharmaceutical products had gained regulatory approval, and around 75% of global
sales were accounted for by only 15 of these (Hopkins et al. 2007). The anticipated stream
of biotechnological medicines has not yet been realised. Instead, biotechnology has
emerged as an incremental, rather than a revolutionary, advance (Nightingale and Martin
2004) in the face of a range of material as well as regulatory and social challenges.

26
The emergence of pharmaceutical biotechnology has involved the establishment of new
relations between the places, biologies and governance of medicine. In Invisible Frontiers,
Hall (2002) describes the ‗race‘ to clone human insulin. Although it does not elaborate on
them, Hall‘s popular science narrative brings to the fore important characteristics of
biopharmaceutical research and development. It emphasises the expert nature of
pharmaceutical biotechnology, particularly compared to the extractive heuristic of drug
discovery. It also highlights that the eventual triumph of the Genentech group was shaped
by the political and geographical context in which early recombinant work was conducted.
In particular, he recounts the key role of containment in the race to synthesise recombinant
human insulin. His account of restrictions imposed on the Harvard team when working in a
containment lab at Porton Down, Britain‘s biological weapons research laboratory,
describes the role of the physical and procedural spaces of the facility:

―Merely entering the P4 lab was an ordeal. After removing all clothing, each
researcher donned government-issue white boxer shorts, black rubber boots, blue
pyjama-like garments, a tan hospital-style gown open in the back, two pairs of gloves,
and a blue plastic hat resembling a shower cap. Everything then passed through a
quick formaldehyde wash. Everything‖ (Hall 2002:254)

Combined with air-locked doors and Byzantine procedures for obtaining access to other
parts of the building, these precautions protected against the contamination of the world
outside the laboratory, but also restricted the progress of research within it. The need to
work in a contained lab was dictated by regulatory requirements for working with human
genes. However, the spaces of biopharmaceutical manufacture which restricted the research
of the Harvard team are co-produced with the natures of pharmaceutical biotechnology. As
Hall (2002) points out, researchers at Genentech in California were simultaneously working
at a lower containment level because their insulin, although structurally identical, was
synthetic and thus outside regulatory concern.

The restrictions placed on biopharmaceutical production aimed to prevent contamination of


products and the environment, and involved moving away from the spaces of whole animal
bodies to those of cell cultures. As such, they involved the elaboration of new production

27
geographies of pharmaceuticals. The post-insulin development of biopharmaceutical
manufacture is described by Michael Kamarck, senior vice-president of Wyeth Biotech:

―Over the next decade, biopharma companies continued to struggle to meet high
demand. Most lacked the manufacturing plants, bioreactors and other equipment
needed to make sufficient amounts of biopharmaceuticals. Briefly put, they needed
stainless steel.‖ (Kamarck 2006:504)

While recombinant biopharmaceutical production allowed an escape from the restrictions


of biological medicines, it was itself restricted by the biofermenters which house E.coli or
CHO cells. In the same way that extractive botanical medicines required botanical gardens
in the 18th and 19th century, so 21st century biopharmaceutical cell cultures require steel
bioreactors, large fermentation tanks whose media and conditions are closely monitored
and controlled to achieve the quality characteristics required of a ‗Good Manufacturing
Practice‘ system. As Morris (2003) describes, the problems of variability and
contamination highlighted by early biological medicines continue to the present.

As a review from the UK National Institute of Biological Standards and Control describes,
for biological medicines the process ‗is the product‘ (Longstaff et al. 2009), and its control
is paramount in ensuring biopharmaceutical quality and safety. The approval of
biopharmaceutical products is based on the verification of this process. In the EU, the
grounds for this are laid out in the regulations summarised in Figure 1.3.

Figure 1.3 European Regulations and Directives relevant to Pharmaceutical


Biotechnology
EU Regulation EEC 2309/93 establishes centralised regulation for biopharmaceuticals and
a European Agency for the Evaluation of Medicinal Products (EMEA). Regulation
726/2004 updates this, laying out procedures for the authorisation and supervision of
medicinal products for human and veterinary use and maintains a centralised authorisation
procedure for biotechnological medicines

Directive 2001/83/EC on human medical products (amended by 2004/27/EC) details


requirements for marketing authorization of human pharmaceutical products.2

2
In December 2008, the EU released a ‗Pharmaceutical Package‘, proposing three legislative changes to
improve access to reliable information, to strengthen the safety monitoring of medicines and to protect EU
citizens from fake medicines. These have not yet been incorporated into EU pharmaceutical governance and
are not considered in this thesis.
28
Standardising and maintaining the unruly natures of biological pharmaceuticals has
restricted their applicability beyond Western pharmaceutical infrastructures. As Kamarck
(2006) continues, building a new manufacturing plant requires five years and hundreds of
millions of dollars. As such, it has to be constructed long before clinical trials demonstrate
the safety and efficacy of the drugs. Moreover, once built, a bioreactor plant cannot easily
be extended.

Facilities for cell cultures are costly to establish and validate, and production is
concentrated in Europe and the United States. Consequently, Morris (2003) suggests the
priorities of the biopharmaceutical industry currently reflect only developed, rather than
developing, world medical needs.

Morris‘s point can be illustrated by the case of antibodies to rabies, provided after a bite
from an infected animal. There are two types of rabies antibodies – human, derived from
biotechnological manufacturing processes, and equine, extracted from horse brains. These
represent both the new and old biological heuristics of pharmaceutical development. The
World Health Organization strongly advocates the use of human antibodies prepared in
cell-culture. However, it acknowledges that these are in short supply and are ―available
mainly in industrialized countries‖ (WHO 2007) due to the concentration of
biopharmaceutical manufacture. Moreover, the requirement for a ‗cold chain‘ for the
delivery of vaccines hinders the mobility of pharmaceutical products. For example, rabies
antibodies produced in cell culture have to be stored at around 2°C. The natures and spaces
of biopharming have been presented as a solution to these challenges.

In the preceding sections, I have introduced the pharmaceutical background to


biopharming, and the limitations of pharmaceutical biotechnology. This snapshot of the
early days of pharmaceutical biotechnology introduces themes that will re-emerge
throughout discussion of biopharming: the relationship between natural and synthetic
biologies; the importance of the places of biotechnology; and the relationship between
technoscientific and social orderings, in this case in the form of regulatory frameworks and
industrial structures. There has been little discussion of the role of publics in
pharmaceutical biotechnology. Yet, in a review of the economic potential of biotechnology,

29
van Reenen suggests that ―perhaps the main problem facing the industry is the public
backlash against GM food and animal testing‖ (2002:123). I explore the relationship
between pharmaceutical and agricultural publics further in section 1.5, but first describe
how the development of agricultural biotechnology over the last 30 years has paralleled its
pharmaceutical counterpart.

1.4 Agricultural Biotechnologies: Plants and Publics

Biopharming represents the ‗third generation‘ of agricultural biotechnologies (Spök et al.


2008). Previous generations have been concerned with traits such as herbicide tolerance
and insect resistance (first generation), and improved food and/or feed quality (second
generation). As in the case of pharmaceutical applications, the development of
biotechnologies has been profoundly intertwined with political, economic and social
histories and geographies.

In the second half of the 1990s the stabilisation of biotechnology achieved by product,
process and programme framings unravelled (Jasanoff 2005). As controversy around
agricultural biotechnologies emerged, their acceptability came to rely on the
trustworthiness of institutions and the social relations of the technology as much as on
scientific assessment of risks.

Jasanoff suggests that today it is impossible to develop a satisfactory model of democracy


―without looking in detail at the politics of science and technology‖ (2005: 6). In the case
of agricultural biotechnology, these politics have been typified by the critical engagement
of European publics. From the introduction of the first genetically modified crops in the
mid-1990s, to the 2008 protests over Monsanto's MON810 maize, public concerns have
been central to the governance of the technology, and in the UK especially, have prompted
a reconfiguration of the relationship between science and society. I discuss these in the
following section, but first introduce the evolution of debates around genetically modified
crops.

30
The complicated story of genetically modified crops and their place in contemporary
agriculture, can best be introduced through the example of a single crop. For Whatmore
(2002) this crop is soya. She charts its ‗hybrid geographies‘ as it moves from prehistoric
domestication to contemporary consumption. However, in introducing biopharming‘s
context, and the complex relations between agricultural biotechnology, existing foods and
its social relations, a better example is provided by maize. Maize has been an important site
for the elaboration of hopes and fears around first generation agricultural biotechnology.
Furthermore, as I explore in detail in Chapters Four, Five and Six, the agricultural and food
characteristics of maize are central to discussion of biopharming.

Since its domestication in Mexico more than 9000 years ago, maize has become one of the
mainstays of global agriculture, capable of growing in a range of latitudes and conditions.
In the 16th century, the farming of maize enabled European colonists to extend their
colonisation of the Americas, while in the early 20th century the development of high
yielding, but sterile, hybrid corn allowed the ‗uncoupling‘ of seed as ‗seed‘ from seed as
‗grain‘ and provided the foundation for the capitalisation of agriculture and the emergence
of the US seed industry (Kloppenburg 2004). As Pollan (2007) describes, processed maize
(corn) products are now found in so many aspects of the modern diet that Americans are
essentially ‗processed corn, walking‘ (2007: 23).

Unsurprisingly then, maize has been at the heart of the development of agricultural
biotechnology. In 2007, Clive James of the ISAAA estimated that 77% of US maize was
transgenic, featuring one or more herbicide tolerant or pest resistant traits (James 2007).
Maize represented 31% of global GM acreage, and was the fastest growing crop. Moreover,
it represented the only GM crop to be grown in Europe.

Maize farming is rare in the UK, where it is grown mainly as forage. However, it makes up
about 14% of all the crops planted in the European Union and 8.5 million hectares were
planted in 2007 (in contrast to 37.9 million hectares in the USA). Of this, around a quarter
was in Romania, with Italy, Hungary and France other important producers (EU 2009). In
Europe, seven EU countries grew transgenic varieties in 2007 (James 2007). However, only

31
111,000 hectares were cultivated across the union, and of this over 75,000 were in Spain
(EU 2009).

While genetically modified crops were easily incorporated into US agriculture, in the UK
and the rest of Europe, their introduction has been less smooth. From 1998 to 2003 a de
facto moratorium on the commercialisation of genetically modified crops was held across
Europe, and even since then the authorisation of new varieties has been hesitant.
Regulatory innovation resulted in the regulatory framework described in Figure 1.4.

Figure 1.4 European Regulations and Directives relevant to Agricultural


Biotechnology

EU agricultural biotechnology regulatory framework established by Directive


90/219/EEC on the contained use of genetically modified micro-organisms and
Directive 90/220/EEC on deliberate release. The latter was replaced by EU Directive
2001/18/EC on Deliberate Release.

European Parliament Regulation (EC) No 178/2002 establishes a European Food Safety


Authority, part of whose remit is to provide scientific advice on genetically modified
foods

The basis for approval of GM foods laid out in EU ‗food and feed‘ and ‗traceability and
labelling‘ regulations 1829/2003 and 1830/2003.

In the UK, the late 1990s saw the emergence of significant public opposition to genetically
modified crops, prompting a three-pronged government consultation in 2003 on the
economics, and science of agricultural biotechnology and in the form of the 'GM Nation?'
consultation on public attitudes. Although this exercise created the possibility for new
forms of societal deliberation around GM (Jasanoff 2005), the public strand was criticised
for a half-hearted and poorly resourced approach, and for methodological flaws (Horlick-
Jones et al. 2006). Nevertheless, all three strands emphasised that a number of scientific
questions remained to be satisfactorily answered.

However, these consultations failed to bring the GM debate to a close, not least because the
UK government appeared to pay little attention to them. In March 2004, Bayer's Chardon
LL herbicide tolerant maize, for use in cattle feed, became the first GM crop to be approved

32
for cultivation in Britain for five years, despite the reservations of all three strands of the
GM consultation, and in particular in light of the significant opposition to GM displayed in
the GM Nation? debate. As Jasanoff describes:

―this product-focussed decision left out of account the message flowing in from many
quarters: that the British public, when consulted through relatively open channels,
did not want to adopt a technology with uncertain benefits‖ (2005: 130)

Recognising the apparent importance of the decision, a spokesman for Bayer CropScience
told the BBC that the decision was "symbolic" and had been made "in the face of a lot of
hostility" (BBC 2004). However, Bayer quickly withdrew its application, claiming that in
light of regulatory restraints, production would be "economically non-viable", while the
government concluded that GM crop cultivation in Britain was unlikely for the "foreseeable
future".

Five years on, the GM debate persists, as demonstrated in 2008 in the case of Monsanto's
MON810 maize, marketed as 'Yieldguard', and authorised in the EU since 1998. The
MON810 variety is the second most approved by worldwide regulatory agencies, and forms
a significant portion of the 18% of authorised GM varieties containing pest resistant traits
(James 2007). The majority of genetically modified crops to date have been engineered for
either herbicide tolerance, such as ‗Roundup Ready‘ varieties, pest resistance, or both. Pest
resistant varieties frequently make use of a gene from the soil microorganism Bacillus
thuringiensis (Bt) which codes for an insecticidal toxin. Bingham (2008) describes the
mobilising promises that accompanied the introduction of the first Bt maize:

―Bt corn… was sold as the answer to the ‗ravages‘ (advertising material) of the
European Corn Borer moth, whose larvae ‗devastated‘ (advertising material again)
around 7% of the Midwestern US corn crop each year (including 20% of some
fields). Farmers were told by promotional literatures that Bt corn would ‗simplify‘
their lives by ‗purifying the environment‘‖ (Bingham 2008:114)

Following Jasanoff (2005), Bingham (2008) describes the introduction of Bt maize as part
of a ‗socio-technical experiment‘ whose apparent immediate success in capturing 20% of
the US corn market served to bolster an overall narrative of technological optimism around
agricultural biotechnology.
33
In 2005 MON810 became the first GM variety to be planted in France, for export to the
Spanish animal feed market. Clive James describes how, in light of the strength of
opposition to GM crops, ―the growing of even a token hectarage of Bt maize in France is an
important and symbolic development‖ (James 2007:84). However, in January 2008 the
French government issued a nationwide ban on cultivating MON810 maize because of
"serious doubts" as to its safety. Austria, Hungary and Germany followed suit. They
activated a 'safeguard clause' in Directive 2001/18/EC, introduced in response to consumer,
NGO and government concerns around GM to regulate the 'deliberate release' of
genetically modified organisms into the environment. According to Article 23, a member
state may suspend planting approval if it has been presented with new scientific evidence
about its effects on the environment or human health. However, in November 2008 the
European Food Safety Authority (EFSA), established in 2003 to provide scientific advice,
concluded that the ban on MON810 maize was "scientifically unfounded"3.

The contested safety of MON810 echoes earlier debates around the toxicity of Bt pollen.
The progressive narrative around Bt crops described above was challenged by the apparent
discovery by Losey et al. (1999) of the toxicity of Bt maize pollen to Monarch butterfly
caterpillars (Bingham 2008). Losey‘s work seemed to provide critics with the necessary
proof of the environmental risk of agricultural biotechnology, as well as an iconic image in
the form of the Monarch, a symbol of US 'Nature'. In response, Monsanto adopted an
argument traditionally deployed by environmental NGOs against regulatory and
governmental analyses of claims of safety, and suggested that the research represented a
mistaken extrapolation from laboratory science to field situations (Jasanoff 2005).

In each case, ‗sound science‘ proved insufficient to answer questions about unknown risks
of agricultural biotechnology. This is indicative of the wider GM debate, where scientific
and governmental assurances of safety have failed to acknowledge both the inherent
uncertainty in such analyses and to open up the assumptions and values underpinning the
development of the technology to wider debate (Wynne 2001).

3 EFSA: ban on cultivating MON810 maize in France is unfounded, GMO Compass 1/11/08 (www.gmo-
compass.org/eng/news/390.efsa_ban_cultivating_mon810_maize_france_is_unfounded.html)
34
The introduction of agricultural biotechnology involved the reworking of relationships
between science and society in Europe, and brought to the fore existing concerns around
the sufficiency of scientific advice and the relationship between science, government and
industry. Moreover, both the rhetoric and the technologies of GM involved new
relationships between developed and developing world, between the natural and the
artificial and between ‗social‘ and ‗scientific‘ space (cf. Babusiaux et al. 2003).

The economic and environmental promise of biotechnology was accompanied and


supported by rhetorical efforts to link GM to earlier agricultural innovations. In 1998,
Monsanto‘s adverts controversially claimed that "Worrying about starving future
generations won't feed them. Food biotechnology will". The positioning of agricultural
biotechnology as a ‗gene revolution‘ or ‗doubly green revolution‘ to repeat the growth in
productivity of the ‗green revolution‘ of the 1960s formed part of the early promise of
biotechnology, and has re-emerged in recent years (Brooks 2005; Vallely 2009). A 2009
Monsanto advert in the New York Times forges new links with climate change, asking: "9
billion people to feed. A changing climate. Now What?" and answers that ―The world‘s
farmers will need to double food production by 2050. Biotechnology can help‖. These
narratives represent biotechnology as a ‗technological fix‘ with its own inexorable
trajectory (Brooks 2005). However, as Marris et al. (2001) describe, such promises have
been received sceptically by European consumers suspicious of the motives involved in the
development of genetically modified varieties.

The relationship between Western biotechnology and developing world food and
agriculture was further highlighted in November 2001 when the journal Nature published a
report by Quist and Chapela. The researchers, from the University of California at
Berkeley, suggested that DNA from U.S.-grown transgenic corn had become incorporated
into the genomes of local maize varieties in the Oaxaca region in Mexico. As geographer
Kathleen McAfee describes:

―The apparent detection of a few invisible, sub-molecular genetic constructs in corn


collected from mountain farmers‘ fields quickly became a cause celèbre for agro-
biotechnology defenders and detractors alike‖ (McAfee 2003a:19)

35
Alongside controversy over the introduction of sterile ‗Terminator‘ seeds, the pollution of
Mexican maize with ‗foreign‘ DNA fired concerns over the threats posed by agricultural
biotechnology to the integrity of developing world biodiversity, farming and political
orders. The transgenic maize presented a challenge to the genomes of Mexico‘s traditional
maize varieties, and to its ability to uphold a moratorium on transgenic maize in the face of
economic and biological pressure from its northern neighbour.

Although other genetically modified crops, particularly soya and cotton, are of equivalent
economic importance (James 2007), maize was, and remains at the heart of the public and
scientific controversy around GM. At the heart of this is its ubiquity in human and animal
food. Maize has come to focus concerns around environmental and health risks, but also
around neo-colonialism, genetic pollution, biodiversity and control of global food
production. The examples above highlight the interconnection of the geographies, natures
and socio-political orderings of biotechnology. Furthermore, they suggest that these
hotspots of socio-technical diversity involve novel engagements between science and its
publics.

1.5 Biotechnologies' Publics

The development of agricultural biotechnology shows that contemporary relations between


science and society are a case of two-way communication. Hopes and fears for the new
technologies have been propounded and contested in public, and as Jasanoff describes:

―at least in Europe—the prerogative of imagining technological futures no longer


rests with governments alone but must be shared with increasingly knowledgeable
publics. Those publics, moreover, approach the promises of biotechnology with
significantly different ethical sensibilities toward nature and different attitudes
toward uncertainty and responsibility from the industries wishing to commercialize
the new technologies‖ (Jasanoff 2006: 291)

However, although both Jasanoff (1995) and Gottweis (1998) describe German protests
against Hoescht‘s recombinant insulin production facility in the 1980s, such animated
public engagement with pharmaceutical biotechnology has been the exception rather than

36
the rule. Indeed, survey research undertaken in the late 1980s and early 1990s by Durant et
al. suggested that medical research was 'paradigmatic' of positive public attitudes to science
(Durant, Evans, and Thomas 1992:171).

A series of Eurobarometer surveys on biotechnology in 1991, 1993, 1996, 1999, 2002 and
2005 (Gaskell et al. 2005) have suggested that a majority of Europeans encourage and
accept 'red' biotechnology applications that have clear medical benefits for the diagnosis
and treatment of human disease, yet are opposed to agricultural applications. The latest of
these Eurobarometer surveys, conducted in 2005, continues to describe a division between
public perceptions of red and green biotechnology:

―There is widespread support for medical (red) and industrial (white)


biotechnologies, but general opposition to agricultural (green) biotechnologies in all
but a few countries‖ (Gaskell et al. 2005:4)

Although sociologically interesting in its own right, the 'red-green divide' in public attitudes
described by Eurobarometers also has wide-ranging practical implications. In particular, it
has become a widely held belief or ‗myth‘ for both supportive and antipathetic stakeholders
in biotechnology (Marris et al. 2001). As described above, van Reenen (2002) suggests that
the greatest threat to the development of pharmaceutical biotechnology in the UK is that of
an overflow of public opposition to GM crops. This distinction thus has potentially serious
consequences for the development of biopharming, a technology which is ostensibly at the
boundaries of red and green.

I now examine how the red/green divide has been explored further, notably in the Public
Perceptions of Agricultural Biotechnology in Europe (PABE) study. I also consider how
the place of publics in agricultural and pharmaceutical biotechnologies is shaped by
assumptions about their role on behalf of governments and experts, but also by publics
themselves.

The Perceptions of Agricultural Biotechnology in Europe (PABE) study (Marris et al.


2001) combines interviews with biotechnology ‗stakeholders‘ and a comprehensive
qualitative study across a number of European countries. As the largest qualitative, focus
group study of the European publics‘ perceptions of biotechnology, it provides an
37
important point of departure for considering biopharming, and offers a nuanced discussion
of the ‗red/green divide‘.

Marris et al. (2001) argue that ―dominant characterisations of the public…do not capture
the full nature of public concerns, nor do they recognise the social, cultural and institutional
factors shaping those concerns" (Marris et al., 2001: 7). The PABE research demonstrates
that participants' perceptions of genetically modified organisms are based in part on a
questioning of the implicit assumptions, values and visions of key actors. Interestingly, the
PABE study also compares and contrasts the discussion of focus group participants around
agricultural and medical applications of biotechnology, providing an empirical basis to
consider public discussion of biopharming.

Medical applications of the PABE discussion constituted only two of the thirteen
predominantly industrial examples of GM presented to focus group participants ‗relatively
late on in discussions‘. Nevertheless, Marris et al. (2001) describe a number of points at
which a medical ‗ideal type‘ is contrasted with problematic food applications. Marris et al.
argue that participants see no alternatives to medicines. As such the stakes involved in
supporting or rejecting the technology are significantly higher, both in terms of personal
and societal benefits. However, in the case of the potential for genetically modified crops to
contribute to alleviating food insecurity, participants were sceptical. Armstrong et al.
(2006) suggest that public support for medical research relates to a vision of potential
personal benefits and a notion of social responsibility: to reduce suffering, prolong life,
save lives and to provide a better future for all. Participants suggest that due to the ubiquity
of food, the risks of GM fall on all consumers, whereas the risks of medicines are targeted
and short term. Participants can choose whether or not to take medicines, and consequently
can control their exposure to risk. However, as Marris et al. acknowledge, this relies on a
particular model of pharmaceutical consumption.

PABE participants also suggest that medicines are taken after the provision of extensive
product information from a trusted individual, adapted to personal circumstances and on the
basis of continued monitoring. The specialised nature of this information meant that
participants were more willing to accept a lack of transparency. This information is seen to

38
be based on more rigorous and effective safety testing and regulation than that undertaken
for foods. There was also a feeling among PABE participants that this testing continued
after introduction, such that medicines were effectively monitored after introduction and
could be effectively withdrawn should unanticipated harmful effects occur. Moreover, there
was also the potential for unforeseen and unintended beneficial effects. Finally, medical
applications of genetic engineering were associated with high price, high quality
applications produced with the motive of meeting existing needs. In contrast, GM food was
equated with low price and low quality and attempts to create demand.

These ideal characteristics are used by Marris et al. to contest stakeholder beliefs that
differing attitudes to biotechnology are dictated by considerations of direct personal need.
Instead, as also emphasised by Armstrong et al. (2006), the envisaged benefits of
biomedicine accrue to both individuals and society. Marris et al. (2001) illustrate that
positive perceptions of medical biotechnologies rely on a range of characteristics of not
only medical products, but of the role of publics in relation to both them and the institutions
that govern and provide them. While publics in the context of agricultural biotechnology
have been framed as ‗consumers‘ or ‗citizen-consumers‘ (Doubleday 2004), in the PABE‘s
medical ideal publics are explicitly patients.

I will return to the role of these ideal characteristics in framing biopharming in Chapter
Seven, in which I present the conclusions of my research. However, the PABE research
notes that this ideal type exists despite the fact that it ‗seems likely‘ that members of the
public are aware that many medical applications do not meet these criteria. They suggest
that people are ‗generally aware‘ that medicines are often for long periods, that medical
information may fail to acknowledge uncertainties, that some medicines may be overly
pushed by health professionals, and that some patients have been actively involved in
pushing for the use of medicines before testing has been completed (Marris et al. 2001).
Perhaps understandably given the scope of their research, the PABE project stops short of
examining whether awareness of these characteristics does exist, suggesting the need for
further research.

39
It is unclear whether a comparison between the conclusions of the Eurobarometer with
those of the PABE study is possible. In Eurobarometer reports the category of ‗medical
applications‘ has evolved from study to study, including not only pharmaceutical
production, but also genetic testing for inherited diseases and therapeutic cloning. Yet the
‗medical‘ characteristics being described by participants in the PABE study are those of
pharmaceuticals, or medicines, rather than of genetic testing or cloning. Throughout the
thesis, I follow the participants in the PABE study as taking ‗pharmaceutical‘ as
interchangeable with medical. However, where it is relevant I distinguish between
discussion of drugs, pharmaceuticals and medicines and that of direct medical applications
of genetic technologies, which do not form the focus of my research.

For Marris et al. (2001), the contrast between agricultural and medical biotechnologies is
the result of a sophisticated engagement on behalf of publics with not only the institutions
and values of agriculture, but with those of pharmaceuticals. The characteristics of
acceptable medical biotechnology form the basis on which it is separated from agricultural
applications and can be taken on into discussion of biopharming. They demonstrate the
complexity of both public and expert interpretations of both agricultural and
pharmaceutical biotechnology. The new technology must capture the potential, the
knowledge about the product (both expert and lay), and the control that typify lay
discussion of medical biotechnologies. In light of these considerations, I now introduce
biopharming, which transgresses the boundaries between red and green biotechnologies and
forms the main empirical focus of the thesis.

1.6 Biopharming

The development of biotechnologies since the early 1970s has been characterised by
contested narratives of their pasts and futures. For both red and green applications these
narratives are performed through their relationships with the geographies, publics and
qualities of existing foods and medicines. I now explore these themes in introducing
biopharming and the Pharma-Planta Consortium, which forms the specific case study of the
thesis.

40
As described by BIO, biopharming is:

―a new application of biotechnology that turns plants into ‗factories‘ that produce
therapeutic proteins used in biopharmaceuticals‖ (BIO 2006:1)

Biopharming is the production of biological pharmaceuticals in genetically modified crops.


In the rhetoric of BIO, it represents the extension of industrial, instrumental approaches to
nature present in approaches to the use of E.coli for pharmaceutical production.

Proponents of the technology argue that biopharming addresses a rapidly increasing


demand for biopharmaceuticals and provides significant advantages over existing E.coli,
yeast and mammalian production systems (Twyman, Schillberg, and Fischer 2005; Ma,
Chikwamba, et al. 2005; Ma, Barros, et al. 2005; Ramessar, Capell, and Christou 2008). As
described above biological, regulatory and physical limitations present restraints to the
expansion of these systems for biopharmaceutical manufacture. In a paper providing an
‗Expert Opinion‘ on emerging drugs, molecular farming researchers argue that the
requirements for labour, equipment and expensive culture media currently make the
production of only the most valuable proteins economically feasible (Twyman et al. 2005).
In contrast, biopharming offers:

―a world in which any protein...could be produced safely, inexpensively and in


almost unlimited quantities‖ (Ma, Drake, and Christou 2003:794).

Biopharming offers the promise of cheap, easily scaleable, safe pharmaceutical production,
based on what another review describes as ‗harnessing the power of agriculture‘ (Fischer
and Schillberg 2004:v). As well as offering the possibility of large scale production, using
plants removes the risks of human pathogens, prions and toxins that can be found in other
biopharmaceutical manufacturing techniques (Basaran and Rodríguez-Cerezo 2008).

The scalability, safety and simplicity of biopharming contrasts with the risks of
contamination and difficulties of expanding existing biopharmaceutical facilities
encountered when using micro-organisms or mammalian cell cultures. In quantifying the
benefits of biopharming, one study estimates that the cost of producing 1kg of recombinant
protein will be 10 to 50 times less than using E.coli fermentation (Kusnadi, Nikolov, and

41
Howard 2000). Moreover, researchers argue that in comparison with bacterial methods,
plants are better able to assemble and fold complex protein molecules (Basaran and
Rodríguez-Cerezo 2008).

I develop discussion of biopharming throughout the thesis, considering how it is narrated,


how its products are related to those of existing agriculture and pharmaceutical production,
and how it shapes and is shaped by the geographies of biopharmaceuticals. Here I provide a
basis for this discussion, introducing the goals of biopharming, its technical achievements
to date, and the social and regulatory challenges described by researchers. Key events in the
development of biopharming to date are summarised in figure 1.5.

Figure 1.5 Milestones in Biopharming

1989 First plant-derived recombinant antibody produced in tobacco in


California (Hiatt, Cafferkey, and Bowdish 1989)
1992 First plant-derived vaccine candidate – hepatitis B virus surface
antigen in tobacco by Mason et al. (1992) of Agristar Inc., Texas
1995 Secretory Immunoglobulin A antibody produced in tobacco by
Julian Ma and colleagues at Guy‘s Hospital, London (Ma et al.
1995)
1998 Mason and colleagues publish first human trial of the
immunogenicity of recombinant antigens delivered in a raw
transgenic potato (Tacket et al. 1998)
2002 Charles Arntzen‘s ‗edible vaccines‘ named as one of Time
Magazine‘s ‗Inventions of the Year‘
Commercial production of trypsin in maize by Prodigene starts
Prodigene fined following contamination of maize and soya in
Nebraska and Iowa
2003 Field trials of gastric lipase-producing maize produced by Meristem
trampled in France
2004 Launch of EU Pharma-Planta Consortium
2006 Dow Agroscience achieves first regulatory approval in the USA of a
plant derived veterinary vaccine for Newcastle disease in poultry
Bayer Innovation, a subsidiary of the Bayer Group, acquires
German company Icon Genetics
2008 Phillip Morris International invests $15 million in Canadian tobacco
company Medicago

42
The initial development of biopharming can be dated to 1986, when Barta et al. (1986)
showed that tobacco and sunflower tissue were able to produce transcripts of human growth
hormone. Since then, a range of biopharmaceutical proteins have been expressed in plants,
including the recombinant proteins and antibodies described earlier, as well as vaccines
(Basaran and Rodríguez-Cerezo 2008).

Early hopes for plant production platforms formed around the idea that foods could be used
to provide stable vaccines without the need for intravenous delivery systems or cold chains.
Genes encoding viral and bacterial antigens can be expressed successfully in edible tissues
of plants (Mason et al. 2002). Early developments included the production of candidate
vaccines for Hepatitis B in bananas and potatoes (Arntzen, Plotkin, and Dodet 2005). Such
edible vaccines were first proposed by Charles Arntzen in the early 1990s, and were
featured as one of Time Magazine‘s ‗Inventions of the Year‘ in 2002 (Lemonick 2002).
However, as Arntzen described to Time Magazine, and as is considered in more detail in
Chapter Five, the use of food products creates challenges for delivering the correct dosage
of vaccine. Consequently, whole food edible vaccines have been replaced by standardised
‗plant-derived vaccines‘.

The second application of biopharming is in the production of recombinant proteins. As


described above, the ability to produce proteins synthetically, and replace dangerous or
limited animal sources, formed a key part of the potential of the early biopharmaceutical
industry. As in the 1970s, the way is being led by insulin. Insulin produced by Canadian
company Sembiosys in safflower plants started clinical trials in the UK in 2008. The
company claims it could meet the global demand from 15,000 acres of farmland4. In 2009
Sembiosys announced that its insulin had been shown to be biologically equivalent to Eli
Lilly‘s Humulin.

While vaccines and recombinant proteins represent a prominent part of biopharming‘s


promise, the most advanced work involves the production of monoclonal antibodies
(Basaran and Rodríguez-Cerezo 2008). As described above, monoclonal antibodies
represent the largest class of biopharmaceuticals in development. Biopharmed 'plantibodies'

4
www.sembiosys.com/Products/ last visited March 2009
43
(Stoger et al. 2002) have been expressed for a number of therapeutic uses. One of the most
advanced of these is CaroRx, produced by Planet Biotechnology in tobacco, based on work
by Professor Julian Ma of St. George‘s Hospital London. It is a secretory antibody against
the Streptococcus mutans bacteria, which causes tooth decay, and as of 2008 was in Phase
II clinical trials (Basaran and Rodríguez-Cerezo 2008).

Unlike bacterial and mammalian systems, which have clearly defined ‗model systems‘,
biopharming‘s diversity in pharmaceutical targets is accompanied by that in the expression
platforms adopted. For example, biopharming companies Prodigene and Meristem have
focussed on maize to produce trypsin and gastric lipase (for treatment of cystic fibrosis)
respectively, while Sembiosys‘s insulin is produced in safflower. US company Biolex uses
duckweed, while Ventria Bioscience uses rice to produce human lactoferrin proteins. The
picture is further complicated by the use of plant or algal cells in bioreactors by companies
such as Greenovation.

Despite this diversity, the field is dominated by the use of tobacco and maize. These
represent 22.5% and 25% respectively of species currently used in producing material for
clinical trials (based on data from molecularfarming.com). Tobacco has played a founding
role in agricultural biotechnology research, and maize is increasingly important as a
genetically modified crop.

This range of crop species is accompanied by, and often dictates, diversity in the forms of
genetic engineering. In the main, biopharming research continues to concentrate on the use
of stable transgenic lines with modified nuclear DNA. Technically this approach to
biopharming is similar to that of first generation agricultural biotechnologies – to create
stable transgenic plants, strands of DNA are inserted into the recipient plant genome using
either Agrobacterium tumefaciens or a ‗gene gun‘ which fires DNA-covered gold particles
into the cell. From the initial cell transformation, a stable transgenic plant can be developed
which expresses the required gene. However, as for first generation genetically modified
crops, these forms of modification are unpredictable (Basaran and Rodríguez-Cerezo 2008).
The use of plant cells in culture, paralleling existing uses of E.coli and CHO cells, and the
transient expression of proteins, ‗transfection‘, using viruses have emerged as alternative

44
options for the expression of transgenic proteins. It is this latter model that has formed the
basis for recent investments in biopharming by Philip Morris and Bayer.

The outline of biotechnology above described the importance of environmental and health
risks in shaping the development of both agricultural and pharmaceutical applications.
These questions are equally prominent in discussion of biopharming, and were highlighted
in 2002 by the ‗Prodigene fiasco‘.

1.6.1 The Prodigene Fiasco

In the 1990s, Prodigene led the development of maize-based pharmaceutical production. In


October 2002 they announced an agreement with Sigma-Aldrich, a chemicals supply
company, to produce ‗TrypZean‘, a recombinant trypsin produced in Prodigene's
proprietary transgenic maize. Trypsin is a digestive enzyme involved in the processing of
proteins in the body, including insulin, and in the production of recombinant proteins in cell
culture. The agreement seemed to represent a validation of the potential of plant-made
production systems.

However, Prodigene‘s success was short-lived. In December 2002, the US Department of


Agriculture imposed a $250,000 fine against the company for violating the Plant Protection
Act (Fox 2003). Prodigene was penalised for two incidents involving test plots of trypsin-
producing maize in Nebraska and Iowa. In the latter, 155 acres of corn were destroyed
because of the risk of cross-pollination by genetically modified corn grown in a nearby
field. In Nebraska 500,000 bushels of harvested soybeans were found by the USDA to be
contaminated with maize which had been grown on the same plot the previous year,
because the farmer did not weed "volunteer" (unintentional) plants from the field in which
the soy was grown. Combined with costs for destroying potentially contaminated crops, the
fine eventually amounted to around $3 million and bankrupted Prodigene (Spök 2007).

For Jasanoff (2005) the Prodigene case represented a continuation of ‗business as usual‘ for
US biotechnology regulation. Rather than representing a systematic failure of regulation,
the Prodigene fiasco was taken as incentive for better monitoring and enforcement, as

45
described in the response of the USDA Secretary to the potential contamination of food
supplies:

"We have to ensure enforcement of biotech regulations in order to maintain


confidence in the systems and the new technologies. And so, when companies don't
adhere to those rules, we will take action as we did ... with...ProdiGene… We are
continuing to work very closely with FDA to ensure regulatory requirements are
clear and that monitoring of licensees that are issued is effective and efficient.... We
also don't want to stifle growth, but we have to have strong and credible regulatory
systems." (Veneman in Fox 2003:3)

Rather than issuing new rules, Veneman suggests that the problem is ensuring that
companies obey existing ones. However, the following year, the USDA increased the
criteria for permission to cultivate pharmaceutical crops, established a ‗zero-tolerance‘
policy on contamination, and concluded that unlike food or feed crops, pharmaceutical
varieties should be subject to continuous regulatory oversight and are not eligible for
‗deregulation‘ (Spök et al. 2008).

Spök et al. (2008) describe how plant made pharmaceuticals present novel challenges to
regulatory bodies, both agricultural, in terms of a novel use of crops, and medical, as drug
regulators deal with a new production concept. Yet neither captures the technology entirely,
and researchers conclude that there is 'no natural home' for biopharming in regulation (Ma,
Barros, et al. 2005:596). In a switch of metaphor, Spök (2007) suggests that EU regulators
are ‗walking a tightrope‘, and that regulatory innovation is required to accompany
technological innovation.

Moreover, the Prodigene case brought to the fore wider concerns about the relationship
between plant made pharmaceuticals and foods. Larry Bohlen of Friends of the Earth
highlighted this in his comment that:

"If the USDA continues to allow biopharm food crops to be planted, someone is
going to get prescription drugs or industrial chemicals in their corn flakes." (in Fox
2003:4)

The motif of drugs in cornflakes has become a recurrent one in discussion of biopharming
and of the Prodigene case. As a representative of the US Consumers Union described:
46
―Drugs have side effects…they should not turn up in our cornflakes‖ (Halloran in
Roosevelt 2003). While the response of Friends of the Earth and the Consumers Union
continued their opposition to earlier applications of GM crops, the Prodigene case brought
forth new social orderings around biotechnology. For example, the Grocery Manufacturers
of America, while emphasising their support for agricultural biotechnology in general,
demanded the mandatory use of non-food crops in pharmaceutical production and increases
in regulation:

―While GMA and its member companies are supportive of biotechnology's current
and future benefits, we must have stronger regulations that will keep pharmaceutical
crops not meant for human consumption entirely separate from the US food supply."
(GMA in Fox 2003)

In addition, the Prodigene case prompted increased US public engagement with the
governance of agricultural biotechnology. While changes to regulations in 1993 had
prompted only 84 public comments, 847 comments were received on the post-Prodigene
changes, many from concerned citizens and ―individuals not commonly associated with the
agricultural biotechnology debate‖ (Stewart and Knight 2005:530).

The Prodigene case has been a significant event in the evolution of biopharming. As well as
ultimately leading to the bankruptcy of Prodigene, the incident brought to the fore the new
relationships between pharmaceuticals and foods in biopharming. It suggests an incipient
reordering of social and technological regimes around biotechnology that is explored in this
thesis through the example of the Pharma-Planta Consortium.

1.6.2 The Pharma-Planta Consortium

The incorporation of biopharming into existing socio-technical networks of food and


medicine, and the co-production of these networks with the new technology, forms the
focus of the thesis. However, rather than examining biopharming as a whole, I use the case
of the Pharma-Planta Consortium to focus discussion.

Biopharming is concentrated in Europe and North America. While the USA predominates
in terms of individual nations, European research represents 38.4% of the total (Basaran

47
and Rodriguez-Cerezo 2008), and is becoming increasingly important (Spök 2007). This
thesis examines the development of European biopharming through the example of the
Pharma-Planta Consortium (PPC).

As introduced earlier, the PPC is a collection of more than 40 laboratories from 34


academic and industrial institutions5. As well as being a scientific endeavour incorporating
molecular biology, plant biology, immunology and vaccinology, the consortium also
involves experts in risk assessment and IP management. The goals of the consortium are
presented in Figure 1.6.

Figure 1.6 The aims of the Pharma-Planta Consortium (From


www.pharma-planta.org)

• To produce recombinant pharmaceutical molecules in transgenic plants


and develop them through all regulatory requirements, GMP standards
and pre-clinical toxicity testing, ultimately to initiate Phase I human
clinical trials.

• To develop robust risk assessment practices for recombinant


pharmaceutical molecules produced in plants, based on health and
environmental impact, working with regulatory authorities within the EU
as well as public groups to ensure that the production systems are as safe
and as acceptable as possible, and that they comply with all biosafety
regulations.

• To develop and refine new strategies for the expression of recombinant


pharmaceuticals in plants, which can be used on a generic basis for
molecules that are normally expressed poorly.

• To develop and generate transgenic plants expressing a second


generation of recombinant molecules that will be used in future clinical
trials.

5 Listed in Appendix II
48
As the PPC press release describes, the consortium is co-ordinated by Professor Rainer
Fischer, of the Fraunhofer IME Institute, Aachen and Professor Julian Ma, of St. George‘s
Hospital Medical School, London (PPC 2004). Its primary goal is to develop antibodies and
vaccines and to enter a candidate product into first stage pharmaceutical trials by the end of
the five year funding cycle in 2009. This goal of the consortium forms a ‗fast-track‘ which
involves the elaboration of both a product and of European regulatory pathways for the
benefit of those who follow. As such, the PPC represents a movement to open the field for
plant made pharmaceuticals in the European Union.

The aim of the PPC is to develop techniques for the production of antibodies and vaccines
to prevent or treat diseases including AIDS, diabetes, rabies and tuberculosis (Ma,
Chikwamba, et al. 2005). To date, the consortium has demonstrated the effectiveness of an
HIV antibody (2G12) produced in transgenic maize plants, which constitutes the ‗fast-
track‘ (Ramessar, Capell and Christou 2008), but also aims to develop both HIV and rabies
antibodies in tobacco.

As can be seen from the examples presented throughout this introductory chapter, the
choice of both application and production crop represents an important one in constituting
relations between biopharming and food, between public and private research and between
developed and developing world agriculture and healthcare.

The structure of the Pharma-Planta consortium is laid out in more detail in Chapter Three,
which describes my research methodology. The project is divided into six work packages,
aligned along a linear axis from basic research to clinical trials. In the EMBO Report which
introduces biopharming, three main challenges are presented to this linear progression –
intellectual property rights, uncertain regulatory frameworks, and public attitudes to first
generation genetically modified crops (Ma, Barros, et al. 2005).

The case of Golden Rice, a second generation genetically modified crop, demonstrates the
importance of intellectual property rights in the research and commercialisation of
agricultural biotechnology. Echoing the goals of the PPC described above, betacarotene-
enriched ‗Golden Rice‘ promised to revolutionise developing world agriculture by
eliminating Vitamin A deficiency in rice-consuming areas. However, intellectual property
49
restrictions prevented the research of Dr Ingo Potrykus being introduced without a license
from Syngenta, the Swiss biotechnology firm and patent holder. Eventually, an agreement
was reached for the ‗humanitarian licensing‘ of Syngenta‘s intellectual property, although
golden rice has yet to be introduced. In light of these concerns, the PPC includes a ‗freedom
to operate‘ review of the relevant intellectual property, but also a ‗Humanitarian Use
Statement‘, making the outputs of research freely available to the developing world.

The regulations relevant to biopharming are divided between agricultural and


pharmaceutical bodies and are still emerging. Biopharming falls under the EU regulations
for both pharmaceuticals and agricultural products described in Figure 1.3 and 1.4.
However, there is ―no ‗natural‘ home or regulatory body for the entire start-to-finish
responsibility surrounding the regulation of PMP crops and their products‖ (Spök et al.
2008:513). As such, both pharmaceutical and agricultural regulators in the EU have moved
to provide specific guidance on biopharming.

The EMEA published draft guidelines on the environmental risk issues involved in
biopharming in 2004 (EMEA/CHMP/135148/2004), which have never been updated. They
also published guidelines on the quality issues, including safety, of biologically active
substances produced in plants in 2002 and 2006, before finalising them in 2008
(EMEA/CHMP/BWP/48316/2006). These refer specifically to the production of
pharmaceuticals for parenteral administration in stable transgenic plants, and are discussed
in detail throughout the thesis. While a range of expression methods were described above,
both cell culture and transfection methods are excluded. As such the regulation limits the
technological options.

In 2008, the EFSA GMO Panel released a draft opinion on the risk assessment of
genetically modified plants used for non-food or non-feed purposes, both medicinal
products and other plant-made industrial compounds (EFSA 2008). In line with the general
provisions of EC Directive 2001/18, they suggest that the risk assessment of the
environmental release of GM plants used for non-food or non-feed purposes should be
focussed on the evaluation of the specific characteristics of these plants, and be carried out

50
on a case-by-case basis. Each of these guidelines and regulatory documents is explored in
more detail in the empirical chapters of the thesis as they become relevant to discussion.

Both intellectual property and regulatory concerns emerge throughout the thesis,
particularly in terms of ensuring the quality and safety of pharmaceutical products,
preventing environmental damage and establishing the relationship between biopharming
and agricultural biotechnology. However, given the central but divergent role of publics in
red and green fields, I am particularly interested in the connection between these publics
and those of biopharming.

Researchers in the PPC and within wider biopharming frequently acknowledge the
importance of public acceptance of the technology. However, while research in the public
understanding of science has repeatedly emphasised that a central lesson from the GM
controversy is the importance of ‗upstream‘ engagement between researchers and publics
(Wilsdon and Willis 2004; Kearnes et al. 2006; Joly and Kaufmann 2008) there is little
evidence of this in the case of biopharming.

While stakeholders adhere to the ‗myth‘ of a red/green divide in public attitudes, the PABE
study (Marris et al. 2001) suggests that medical applications must meet a range of
characteristics to achieve support. Yet the example of Prodigene suggests that biopharming
may involve shifts in public engagements with biotechnology. Although biopharming
researchers themselves recognise the importance of public attitudes to the development of
the technology (Ma, Barros, et al. 2005), the availability of studies is limited. In fact there
have been relatively little detailed research into public attitudes to biopharming, particularly
in Europe, and the PPC itself has made no effort at public consultation or participation.

Much existing research has focussed on the USA and Canada (Kirk and Mcintosh 2005;
Knight 2006; Nevitt et al. 2003). These have predominantly been survey studies based on
telephone interviews. While the number of comments received on changes to regulatory
requirements following the Prodigene fiasco might suggest a shift, these studies generally
identify support for the technology. The most in-depth North American study of
biopharming‘s publics has been undertaken by Edna Einsiedel and colleagues in Canada. A
modified focus group methodology was used to try and understand 'early-stage' consumer
51
perceptions to aid policy development (Einsiedel and Medlock 2005). This study identified
a number of key concerns around biopharming, including the potential for cross-pollination
and contamination of food crops; issues of safety, regulations, and policing; potential long-
term side effects and dissonance between the perceived interests of those developing and
growing these crops and the public interest. The focus group stage of research was followed
up in 2007 by reconvening a Citizens‘ Panel which had previously met to discuss
agricultural biotechnology in 1998. The 12 member panel discussed the advantages and
disadvantages of biopharming, concluding that despite potential health and environmental
risks, the technology presented overall benefits and opportunities for agriculture. In
addition, an online consultation of a randomly selected group of 400 ‗involved citizens‘
suggested that a large majority (80%) were supportive of the technology‘s development
were regulation to be sufficiently strong (GE3LS Alberta 2007).

However, the European context for agricultural biotechnology has been significantly
different from that in North America. Biopharming, as ‗bio-plants‘ was included in
Eurobarometer 64.3, which linked public attitudes to the state of regulation, asking:

―Another application is the use of genetically modified plants in the production of


medicines and pharmaceutical products. These genetically modified plants will be grown
in enclosed greenhouses and the expectation is that this could be a cheaper and more
efficient way of manufacturing medicines. Which of the following best describes your
views?‖ (Gaskell et al. 2006; my emphasis)

Respondents to this question were asked not for their general positive or negative feelings
but for their approval in relation to levels of regulation. The study found that in all EU
countries except Austria, support outweighed opposition. 25% of Europeans (27% of
British respondents) approve of the use of bio-plants with usual levels of government
regulation, which suggests that these respondents not only feel sufficiently positive about
bio-plants, but also have confidence in government and its regulatory capacity. In addition,
37 per cent approve of bio-plants if they are tightly regulated (44% of British respondents).
Finally 11 per cent (8% of British respondents) do not approve of the technology under any
circumstances (Gaskell et al. 2006). The Eurobarometer would seem to suggest significant

52
support for the technology, based on the presence of novel regulatory structures to
accompany the technology. However, the Eurobarometer question presents a specific
framing of the technology, based on production in contained facilities, and emphasising the
cost and efficiency advantages of the technology. As I explore in the empirical chapters of
the thesis, these conditions are by no means assured. In contrast, qualitative studies of UK
public attitudes have been less conclusively positive.

In its consultative role, the Agriculture and Environment Biotechnology Commission


(AEBC) sponsored a study into non-food applications of agricultural biotechnology,
including the production of an HIV microbicide and monoclonal antibodies to prevent
dental caries. The report concluded that despite the apparent presence of a ‗red/green‘
divide in public attitudes to biotechnology, the majority of people were more cautious about
medical applications than might perhaps have been expected (CWR&D 2005). The report
comments that "despite the strong desire for medical breakthroughs neither of the two
medical application case studies drew sustained support" (2005: 40). Attitudes to both were
related to the perceived benefit and the risks of contamination. The dental caries application
was not well regarded by participants. In the case of the HIV application, the study found
that the potential benefits of a microbicide were seen to be outweighed by the
environmental and health risks of growing GM plants in the field, although support would
have been greater for such growing if the pharmaceutical had offered a cure or viable
treatment, for HIV/AIDS.

These studies provide a context for the current research. However, the purpose of the
current research is not to provide a summary of public attitudes to biopharming6. Instead,
the thesis examines how public and expert perspectives are co-constructed. It considers the
heterogeneous processes through which biopharming is ‗placed‘ among existing
agricultural and pharmaceutical biotechnologies through which new places are produced.
As described in sections 1.3 and 1.4, the development of biotechnology has involved the
production of new places of pharmaceutical production, and new relations between
developed and developing worlds, and between farms and field trials. In concluding this
introductory chapter, I describe the ways in which the changing relationship between
6
Although I have produced such a report as a separate publication (Milne 2009)
53
science and society identified above has been approached from within STS, and introduce
my use of the heuristic of ‗placing‘.

1.7 Relating Science and Society

Hopes and fears for agricultural and pharmaceutical biotechnologies have been produced
alongside a red/green divide in public perceptions, as highlighted by the PABE study
(Marris et al. 2001). The PABE study is part of a ‗contextual‘ tradition in the public
understanding of science, which suggests the importance of local lay knowledges in the
formation of attitudes to science and technology. Since the early 1990s, researchers have
argued for increased attention to the ―interactions between people's existing understandings
of particular situations and those that emanate from science‖ (Wynne 1991:113). Work
from this critical PUS perspective attempts to examine how publics assess scientific claims
to knowledge, and describe how these forms of assessment are embedded in local, and
located, cultural practices and experiences of the everyday (Wynne 1991, 1992, 1996; Irwin
and Michael 2003; Michael 2002). In particular, issues of trust and of social relationships
come to the fore and knowledge of scientific institutions contextualises scientific
knowledges, rather than vice-versa.

The contextual approach emerged in response to and in development from, the ‗deficit
model‘ that dominated early work in the public understanding of science. Irwin and
Michael (2003) describe how work in the public understanding of science has been
characterised as divided into areas informed by two models, 'deficit' or 'positivist' and
'contextualist' or 'interpretationist'. Within the former, it is assumed that a lack of public
support for science is the result of public ignorance, and that this ignorance decreases the
ability to become properly involved in the democratic process. Within the deficit model
therefore, public understanding of science equates to knowledge of science. Although the
deficit model has been strongly criticised for its equation of knowledge with attitudes
(Irwin and Michael 2003), Durant et al., argue that this may be useful in those areas of
science in which there is a consensus, and in which knowledge is therefore relatively

54
uncontested (Durant et al. 1992). However, in the case of biotechnology, such areas are few
and far between.

The contextual model, on the other hand, highlights the existence and importance of 'lay
knowledges‘ or 'lay epistemologies'. The now canonical example of research examining
relations between ‗publics‘ and ‗science‘ in this vein is Brian Wynne‘s study of hill sheep
farming in Cumbria, in which local knowledge of farming conditions was ignored by
mainstream scientific advice to government and farmers about the persistence of
contamination from the Chernobyl nuclear disaster (Wynne 1992). Wynne describes how
assertions from government scientists that contamination would clear in weeks contradicted
farmers‘ own knowledges about the contingencies of farming in the Lake District, and took
no account of the idiosyncratic features of the area. The globalising and homogenising
model adopted by scientists undermined the status of local knowledges, and created
disillusionment with scientists‘ ability to predict and manage risks. The public reception,
perception and use of knowledge are thus not only related to the public‘s formal
understanding of its content, but involve an awareness of ―the forms of institutional
embedding, patronage, organisation and control of scientific knowledge‖ (Wynne 1992:42).
In these terms, ignorance of scientific knowledge is no hindrance to democratic
engagement. Indeed, an overly narrow focus on scientific knowledge may instead lead to
the impoverishment of democratic discussion.

Felt et al. (2007) suggest the emergence of a third approach to the public understanding of
science, in which the role of the public is a more active one, not just in the reception, but in
the production of knowledge. This model has emerged from suggestion that there is a need
to develop a more socially responsible science, and that scientists need ―to find ways of
listening to and valuing more diverse forms of public knowledge and social intelligence‖
(Wilsdon and Willis 2004:1; Kearnes et al. 2006).

Working with this ‗co-productionist‘ model, Jasanoff emphasises how regulatory and
public knowledges emerge from common situated processes. Public life becomes the arena
in which technoscientific claims are produced and elaborated, rather than the site of ‗public
attitudes to…‘. In their contribution to the New Handbook of Science and Technology

55
Studies, Bucchi and Neresini (2008) describe how, rather than juxtaposing expert and lay
knowledges, a ‗co-productionist‘ approach describes how expert and lay knowledges do not
‗encounter each other‘, but are the result of common processes. In the terms of Irwin
(2001), they are ‗co-constructed‘. The relationships between these models, and the roles of
publics within them, are presented in Figure 1.7

Figure 1.7 Three Models of the Science and Society Relationship (adapted
from Felt et al. 2007)

Deficit Model
Science is seen as separated from society. The public thus does not
intervene in the process of knowledge creation.
Technoscience is a source of progress as long as it is well used by different
actors
Mistrust towards science comes from public illiteracy, ignorance and
superstitions
Thus scientists have to instruct and educate the public
Knowledge is globally applicable
Contextual Model
Science has to be opened to debate to enrich it (but the public does not
participate in the creation of scientific knowledge)
Frontiers between specialists and non-specialists are blurred
The legitimacy of decisions comes from open debate
Scientific knowledges co-exist with local knowledges. Each is situated in
particular circumstances of production
Co-production of Knowledge
Science is closely intertwined with society
Citizens and ―concerned groups‖ get actively involved in the process of
knowledge production of direct use for them
Knowledge created in laboratories is central, but it is created through
interaction with citizens

56
Michael (2002) highlights commonalities between existing approaches to the public
understanding of science (PUS). Michael suggests that each involves a humanistic
understanding of publics which limits them to either ‗comprehension‘ or ‗apprehension‘.
Instead, he asks

―what happens to PUS when one reconstructs this member of the public so that its
constituents include nonhumans? That is to say, where the public is conceived as a
disparate collective that incorporates not only humans in social relations but also
technologies that in part mediate those very social relations‖ (Michael 2002: 358)

In answering this question he draws on Whitehead‘s concept of embodied ‗prehension‘ or


‗grasping‘ to develop a model of the public as emergent from associations between diverse
elements. Within this heterogeneous PUS ―we would seek to map how science is prehended
not only through representation but also in everyday material encounters with, for example,
technology and nature‖ (2002: 373). Similarly, in her introduction to the ‗idiom‘ of co-
production, Jasanoff (2004) stresses that there is a constant intertwining and interaction
between the cognitive, material, social and normative features of new science and
technology. She suggests that co-production is not just about ideas, narratives and rhetoric,
but also about new materialities - new concrete physical things. In this sense, studying what
Michael terms the heterogeneous public understanding of science involves recognizing and
capturing the ways in which what makes up the human, the social and the natural, or the
medical and agricultural are ‗tied to‘ each other (Michael 2000).

For example, the story told by Wynne (1992) could also be read as pointing to the
importance of the material characteristics of ovine biologies, of radiochemistry and of soils
in shaping engagements between researchers and farmers. Moreover, the relationship
between farmers and researchers in Wynne‘s case study is circumscribed by spatial factors,
by both the location of their encounter in the Cumbrian hills and the relationship of this
place with placeless scientific models of caesium removal from soil.

In a similar vein, Law (1992) has suggested that ―people are who they are because they are
a patterned network of heterogeneous materials‖ (1992: 3). Irwin and Michael introduce the
concept of the ethno-epistemic assemblage (EEA) to capture how scientific and social
change is co-constructed (Irwin 2001; Wynne 2002), but also how novel technoscience is
57
embedded in existing networks of mundane technologies and everyday social interaction
(Michael 2006).

The concept of an ethno-epistemic assemblage does away with an a priori contrast between
expert and lay perspectives, replacing this with the notion of conflicting assemblages
characterised by heterogeneity and fluidity, and composed of diverse knowledge and
resources, including but not limited to scientific knowledge (Horst 2007). The concept of
assemblage implies the formation of transitory communities ―that coalesce and then melt‖
(Irwin and Michael 2003: 108). In placing expert and lay perspectives alongside one
another and investigating how they come together and diverge around various material and
discursive features of biopharming, the current thesis adopts such an approach.

EEAs capture the processes of co-construction of the lay and expert and the natural and the
social. For example, Irwin and Michael (2003) highlight how actors involved in
technoscientific discussion co-construct multiple 'natures', drawing on resources which
circulate between communities, institutions and organisations. As they describe in drawing
on Michael and Brown‘s work on xenotransplantation, rather than being 'largely scientific'
or 'largely lay', there is 'discursive promiscuity' between groupings. The heuristic of the
ethno-epistemic assemblage allows us to examine how such groupings come to hold
together, "expanding the range of entities, actors, processes and relations that get blurred
and mixed up" (2003: 114).

The HPUS/EEA model also extends the focus of critical PUS to situate it in relation to non-
local knowledges and practices, and emphasising the ways in which multiple
interconnected locals are assembled into the global. While Wynne‘s example emphasises
the importance of local knowledges, Michael and Brown (2005b) argue that, public
understandings of science have ―been conceptualized as overly local‖, and that far from
being purely local, ―lay local communities are shot through with the complexities and
contradictions that typify such contemporary dynamics as globalization and consumerism‖
(2005b: 42). Publics and scientists are both embedded in distinctive local cultural contexts
and able to draw on a globally accessible culture, blurring distinctions between the local
and the global. Similarly, local places can no longer be considered as bounded, settled and

58
stable, but as points at which multiple connections and interrelations come together (Jess
and Massey 1995).

1.8 Places and Placing

The question of place, and the relationship between global and local circumstances and
circulations of foods and medicines, is one that has emerged throughout this introduction,
and which I pursue throughout the thesis. The language of place permeates discussion of
biotechnologies in general and biopharming in particular. However, within science studies
and the public understanding of science, questions of place have often been overlooked.

As described above, researchers suggest that biopharming has no ‗natural home‘ (Ma,
Barros, et al. 2005). The choice of a regulatory ‗home‘ is an interesting metaphor. As
geographer Gillian Rose describes, a home is a meaningful location through which and
within which identities are constructed (Rose 1995). However, the vocabulary of place is
not restricted to biotechnology researchers. Jasanoff (2005) draws on similar metaphors in
describing, first generation agricultural biotechnology was:

―never viewed in isolation from wider issues of agricultural practices, nature


preservation, and the integrity of food. These [GM] products had to take – or, more
accurately, make – a place for themselves on tables and supermarket shelves, as
harbingers of a new mode of agricultural production‖ (Jasanoff 2005:120; my
emphasis)

For subsequent ‗generations‘ of agricultural biotechnology, the making of places described


by Jasanoff becomes qualitatively more complex, and involves the re-placing of existing
relationships.

Michael (2006) describes how technoscience is ‗located‘ in everyday life: in laboratories,


media representations, standards, or in mundane artefacts and expertises, as well as in the
publics it constitutes. Michael‘s description is useful in drawing attention to the diversity of
sites at which technoscience is manifested in the everyday circulations of foods and
pharmaceuticals that I chart in this thesis. I aim in this discussion to draw attention to the
multiple ways in which these locations are accomplished, not least by bringing in work in
59
human geography which I introduce in Chapter Two. However, in bringing these
approaches together, I move away from the vocabulary of locations and locating. Instead,
one of place and ‗placing‘ captures the establishment of spatial but also material and
discursive relations between experts and publics, agriculture and pharmaceuticals.

As Staeheli (2003) describes, place incorporates location, but also extends beyond it. She
provides a breakdown of the uses and meanings of place in human geography. She
examines the conceptualisation of place as: physical location; social location; context;
constructed over time; and as process.

Staeheli (2003) suggests that the first of these is that of place as 'material, grounded and
bounded', and as contrasting with abstract space. The second, that of social or cultural
location, captures the meaning used by Douglas in her description of matter 'out of place'
(2002). In this sense, place represents a sense of 'belonging' and conversely of
transgression. Thirdly, Staeheli describes place as context. It is perhaps this meaning that
has traditionally been most prominent in STS, particularly in laboratory studies, or in
Latour (1983) and Callon's (1986) early descriptions of actor-networks in Paris or St
Brieux. She then describes how places are conceptualised as socially constructed and
dynamic. Place is thus emergent over time and continually shaped and re-shaped. Jasanoff's
(2005) description of 'making a place' for agricultural biotechnology might be seen in these
terms.

The final conceptualisation described by Staeheli (2003) is that of place as a process.


Thinking about place as an active process of becoming, as ‗placing‘, brings together the
interconnections between localities, the construction of place and the physical and social
locations of the other definitions. It is this combination that I attempt to capture through the
use of the device of 'placing' in this thesis. Moreover, this captures the emergent nature of
places and the ‗placing‘ of biopharming within the narratives of agriculture and
pharmaceuticals.

Placing involves situating and ordering, not only spatially but also temporally and
relationally – as the OED defines it, to place is to class, to date and to locate (OED 2008). It
is a process through which novel technoscience can be located, through which it is
60
classified in relation to both quotidian and exotic ‗things‘, and through which it can be
inserted into a narrative of technological and scientific development. However, as work on
co-production suggests, it also involves the elaboration of new places, new relations and
new narratives. By foregrounding the placing of biopharming in this way, I contribute to an
enhanced conception of place within STS, and highlight its role in the realisation and
contestation of the technological promises with which I started this chapter.

Building on the discussion of place and biotechnology introduced in this chapter, I explore
two questions in this thesis. Firstly, I ask how the promise of biopharming is performed
through, and shaped by, the technology‘s relations with existing green and red
biotechnologies. Secondly, I explore how the heuristic of placing can be used to explore the
heterogeneous ways in which these relations are established by experts and publics. I
consider how the elaboration of biopharming's futures is both a discursive and material
accomplishment, and extend this discussion by considering how technological futures are
produced and challenged through the 'places' and geographies of biopharming.

1.9 Thesis Outline

In this chapter I have introduced biopharming and situated it within a context of increased
dialogue between science and society, in which natural and social orderings of the world
are produced alongside each other. In particular, I described how the development of both
pharmaceutical and agricultural biotechnologies can be understood only by examining the
heterogeneous socio-technical processes through which they emerge. In the remainder of
the thesis I examine how expert and public perspectives on biopharming are produced
through the same processes, and structure my discussion around three ‗placings‘ of the
technology.

In Chapter Two, I introduce relevant literature from STS and geography to consider how
the co-construction of science and society does not occur at a single site, but through the
discourse, materials and spaces of science and technology‘s engagements with the everyday
world. In Chapter Three, I introduce the methods used to study expert and lay placings of

61
biopharming. I consider the merits of a qualitative approach, and both the theory and
logistics of the focus groups and interviews used in the research.

The main body of the thesis is divided into three empirical chapters which together chart a
trajectory of biopharming from imagination to realisation. While the order of these chapters
suggests a linearity, they describe discussions and practices that are overlapping and
iterative. Thus the imagination of futures and pasts of biopharming, described in Chapter
Four, shapes the materials and places described in Chapters Five and Six, but these also
drive a reconsideration of these imaginations. I emphasise that expectations and promises
around biopharming must be understood within particular agricultural or pharmaceutical
narratives, and through their interaction with the obdurate matters, natures and geographies
of agricultural and pharmaceutical production.

Chapter Four introduces biopharming‘s place in time as a programme of medical research. I


consider how expert and lay actors combine hopeful pharmaceutical futures and heroic
medical histories with troubled and sceptical agricultural pasts and promises. I describe
how the PPC‘s version of biopharming, and support among the publics researchers, relies
on its position within imagined geographies of disease and of agricultural production, and
how these in turn shape the technical choices made by biopharming researchers.

While Chapter Four concentrates on the temporal placing of biopharming in relation to its
pasts and futures, Chapter Five examines how the promise of biopharming is encountered
and established as its products, plant made pharmaceuticals, are placed among, and related
to, existing medicines and foods. I consider how differing relationships between the form
and effect of pharmaceutical and food products contribute to regulatory and vernacular
classifications of their quality and safety.

Chapter Six continues this discussion of the quality and safety of pharmaceutical and food
products to explore how biopharming‘s promise is placed among food and pharmaceutical
production. I consider the relationship between these production processes and their places,
extending discussion of the role of geographies in shaping biotechnology described in
section 1.3 and 1.4. I examine how, as biopharming is ‗placed‘ in a more narrowly

62
geographical sense, relations between it, agriculture and pharmaceutical production
concresce.

Finally, in Chapter Seven I bring together the discussions and conclusions emerging from
each of these chapters. I consider what has been learnt about the multiple roles of place in
the relationship between biopharming and existing biotechnologies, foods and medicines. It
provides suggestions for the future direction of research in the geographies of science and
the sociology of expectations, and for the development of biopharming itself.

63
CHAPTER 2: OF PLACING AND PROMISING

Biopharming inherits the promises and problems of previous generations of biotechnology,


and is co-produced with new configurations of the natural and the social, the expert and the
public. To capture these, I make use of the device of ‗placing‘. I intend it to emphasise the
role of the spatial and geographical features of technoscience, but also to describe the
importance of belonging and continuity, as well as novelty and discontinuity, in the
introduction of novel technologies.

In this chapter I consider the contribution of existing work to understanding the temporal,
relational and spatial placing of biopharming, and the relationship between these and
potential technological futures. The diverse yet connected approaches presented in this
chapter represent a theoretical basis from which to examine how biopharming is ‗placed‘ in
relation to the diverse characteristics of agricultural and medical technologies and
biotechnologies.

Narratives of biotechnology‘s pasts and futures have been central to expert and lay
understandings of their development. To address the role of these narratives in the case of
biopharming, I turn to work in the sociology of expectations, which describes the
performative and organising role of futures in technological innovation. Moreover, this
work also emphasises the relationship between the materials and artefacts of novel and
exotic technologies and those of mundane everyday life. However, in my discussion of
pharmaceutical and agricultural biotechnologies, I described the importance of questions of
place. The emergence of the new technologies occurred alongside the elaboration of new
places of production and regulation. To consider the relationship between the places of
biotechnology and its promise further, I bring work in the sociology of expectations
together with that in the geographies of science.
2.1 From Efficacy to Affectivity

A burgeoning body of research in STS focuses on the ‗sociology of expectations‘. It


explores the role of promising futures in shaping new technologies, and emphasises the
performative role of these futures in the stabilisation of stable socio-technical networks
around new technologies and biotechnologies. As such, it highlights the role of the
imagination in positioning novel technologies in relation to existing ones, in inserting new
technologies into narratives of technological and social development, and in constituting
new relationships between experts and publics. In the discussion below, I describe how
notions such as ‗communities of promise‘, ‗imagined communities‘ and ‗imagined
geographies‘ contribute to understanding the relationship between public and expert
understandings of future promise and past success.

In setting out a programme for the sociology of expectations, Brown and Michael (2003)
suggest that:

―on the one hand, new technologies emerge in the context of mundane and
unremarkable networks of established actors (regulatory, economic, scientific,
public). On the other hand, they must de-stabilize these networks in order to establish
their own amenable and fertile associations‖ (2003: 14)

In focussing on expectations and future visions, this work emphasises the importance of
narratives in these de-stabilisations and re-associations. In Chapter One, I introduced
Jasanoff‘s influential study of biotechnology as an example of the ‗placing‘ of
biotechnologies. Jasanoff (2005) emphasises the role of frames and framing in the
divergent responses to biotechnology of the three political cultures she studies. While the
‗technological frames‘ described by Bijker (Bijker 1995) are resolutely heterogeneous, in
the more traditionally sociological terms adopted by Jasanoff (2005), framing is a process
of imagination and narration through which novel objects, events or experiences are
understood and narrated in terms of existing experiences and understandings (Goffman
1974; Jasanoff 2005). Frames allow discipline and order to be imposed on unruly events by
relating them to wider social narratives. As Felt et al. introduce, societies make use of such
narratives to express wider imaginations about the world. These narratives tacitly define

65
what action is possible and acceptable, and project and impose classifications and ordering
(Felt et al. 2007:73).

What Jasanoff highlights effectively is that biotechnology becomes framed in different


ways for different national regulatory actors, and that the salient aspects of the
technological innovation are not the same in each context. The adoption of a comparison
between national contexts demonstrates how biotechnology is framed by ‗local‘
contingencies, including national legal codes or styles of governance and culturally specific
conceptions of the form and role of ‗expertise‘ and ‗the public‘.

The framing approach adopted by Jasanoff is valuable in presenting the variety of meanings
attributed to first generation biotechnologies through their narration. Levidow and Carr
(2007) describe similar risk framings, and suggest that contests between frames involve
representing the dangers and opportunities of biotechnology in ways which favour the
imagination of particular socio-technical futures.

2.1.1 The Imagination and Technological Futures

Imaginations of the future – expectations of technological futures – shape the development


of new technologies. The ‗sociology of expectations‘ describes how visions of the future
development of technoscience are performative in their present (Michael 2000). Yet as
Brown, Rappert and Webster (2000) emphasise, there is a multiplicity of available futures.
Consequently, addressing their role in the development of new technologies requires a shift
in analysis from looking 'into the future' to looking 'at' the future. Visions, promises or
expectations act in the present to shape the research questions, funding commitments and
institutional orderings which drive technologies such as pharmacogenetics (Hedgecoe and
Martin 2003), stem cells (Geesink, Prainsack, and Franklin 2008) or xenotransplantation
(Brown and Michael 2004). The circulation of expectations thus constitutes a space in
which negotiation, challenge and reconciliation around new technologies occurs. The
emergence of these technologies depends on assumptions about their future relations to
other technologies (Brown, Rappert and Webster, 2000) and about users, markets,

66
regulations, and technical progress (Geels 2007). As Martin, Brown and Kraft (2008)
describe:

―expectations extend from and contribute to the stabilization of networks, specific


socio-technical identities and the creation of particular relationships between actors
and within communities based on mutual imagined understandings‖. (Martin, Brown
and Kraft 2008:38)

Brown and Michael (2003) highlight how claims about the future potential of technologies,
particularly as hype, are at their strongest when uncertainty about the future of the
technology is greatest, and that these claims serve to marshal support and establish
particular visions as the technology becomes ‗scripted‘ (Akrich 1992). Importantly for the
study of biopharming in this thesis, technological visions are a key site around which
encounters between expert and public understandings of technologies occur, and through
which public support or opposition is generated. If technological promises are accepted,
they are translated into forceful requirements, directing future activities. Expectations
consequently contribute to the construction of a technological narrative which provides
both a vision of the future and a means and imperative of getting there (Deuten and Rip
2000; Brown 2003).

Hedgecoe and Martin‘s (2003) discussion of pharmacogenetics (PGx) exemplifies the


activities involved in securing specific futures and negotiating a place among existing
technologies. They describe how visions of the future of technologies actively shape their
development. Hedgecoe and Martin (2003) argue that conflicting promissory visions of
PGx help constitute the socio-technical network of its development in its early stages of
uncertainty and become embedded in research agendas and industrial structures. Visions
thereby become ‗forceful‘ (Van Lente 2000) in actively shaping and driving the trajectory
of a nascent technology.

Hedgecoe and Martin (2003) discuss the presence of two visions for PGx, one of increasing
drug efficacy, and a second of drug safety. These visions situate PGx in differing relations
to existing drug development. The second of these concentrates on understanding and
averting adverse drug reactions, placing, or ‗rooting‘ in their terms, modern

67
pharmacogenetics in relation to existing medical practice, making it familiar and
uncontroversial (Hedgecoe and Martin 2003). Hedgecoe and Martin contrast this with a
rhetorically powerful vision of pharmacogenetics which instead presents it as discontinuous
with existing drug development, as representing ―the potential for creating a new type of
medicine‖ (2003: 340) based on the development of new drugs.

Hedgecoe and Martin (2003) argue that a commitment to a particular vision involves not
only a scientific judgement, but also specific research questions, prescribing practices,
strategies for drug development, commercial products, business relationships and
definitions and stratifications of disease. As such, in the terms introduced in Chapter One,
expectations and visions temporally ‗place‘ pharmacogenetics among existing
pharmaceutical technologies.

Brown (2007) describes the role of expectations and imagined futures in early debates
around agricultural biotechnology. As described in Chapter One, the development of both
agricultural and pharmaceutical biotechnologies was characterised by inflated and
spectacular claims. In the late 1990s, in response to the widespread opposition to
genetically modified crops, agricultural biotechnology firms again attempted to mobilise
hopeful futures for their technology. Monsanto‘s adoption of the corporate slogan ‗Food,
Health and Hope‘ and subsequently, ‗Imagine‘ represented attempts to move debate away
from ‗regimes of truth‘ and instead establish the affective and symbolic value of
biotechnology (Brown 2007; Moreira and Palladino 2005).

The examples from agricultural and pharmaceutical biotechnology presented in Chapter


One emphasise that attempts to ‗colonise‘ (Brown and Michael 2003) the hopeful ground
extend widely. However, futures are particularly forceful in the mobilisation of public and
financial support for biomedical technologies (Good 2001). Enthusiasm for the potential of
medical research derives not from existing applications, but from the possibility of future
treatment as we continue to rely on the promise of pharmaceutical research and
development. While faith in research in agricultural biotechnology has been shaken,
support for pharmaceuticals remains high. As Good puts it:

68
―Enthusiasm for medicine‘s possibilities arises not necessarily from material
products with therapeutic efficacy but through the production of ideas, with potential
although not yet proven therapeutic efficacy‖ (Good 2001:397)

Such ideas are part of a ‗political economy of hope‘ based on the circulation of shared
images of the promise of biomedical research, what Good terms the ‗medical imaginary7‘
and Waldby (2000) the ‗biomedical imaginary‘. As Squier (2004) points out, this
‗speculative and propositional‘ imaginary is present not only within biomedicine, but in
wider cultural discourse. It is central to the enrolment of a range of actors and communities
in the hopeful futures of biomedicine and biomedical innovation (Moreira and Palladino
2005; Brown 2007). Becoming aligned or associated with biomedical promise thus shapes
both the form and understandings of emerging medical technologies such as biopharming.

The work of Good, Brown and Moreira and Palladino focuses on medical interventions
such as stem cell banking. However, this shift in emphasis from ‗truth‘ to ‗hope‘ has yet to
be examined in detail for biopharmaceutical technologies. As for other medical
interventions one might expect the value of these applications, including biopharming, to be
constituted through the technology‘s ‗affectivity‘, or its ability to mobilise financial and
social support.

2.1.2 Promising Communities

Studies of expectations have often concentrated on the actors closest to innovation,


particularly researchers and companies. However, the promises of agricultural
biotechnology in particular have been portrayed and contested in public and by publics.
Promises and visions of new technologies tap into socially and culturally embedded
imaginaries of technological development which are open to challenge. In this section I
examine how promises are placed in relation to wider social narratives of technoscience
and how they enable publics and experts to come together in the ‗assemblages‘ or
ephemeral communities described by Irwin and Michael (2003).

7
An imaginary is a set of socially and culturally embedded assumptions. These are not necessarily false, but
exist primarily in the imagination. Marcus (1995) suggests that such imaginaries unwittingly shape future
worlds and possibilities through technoscientific practice and innovation.
69
Martin, Brown and Kraft (2008) suggest that expectations act as ‗mutual imagined
understandings‘ for the stabilization of socio-technical identities. They describe the
formation of heterogeneous ‗communities of promise‘ as the means through which visions
of technological futures are constituted, circulated and exchanged, through which different
groups of actors are enrolled and aligned, and through which particular technological
identities are formed. As Brown (2003) introduces, communities of promise are sites of
exchange among investors, consumers, regulators, researchers and other actors. Within
them, expectations and promises constitute and organise binding relationships (Brown
2003).

Communities of promise are heterogeneous, dynamic and ephemeral (Martin, Brown and
Kraft 2008; see also Irwin and Michael 2003). They form, disband and reform as
technological expectations develop. This dynamic interaction between groups around the
promise of novel technoscience provides a mechanism through which novel technologies
such as biopharming are placed as part of a temporal narrative of technological
development, and whereby the impact of this placing on the material form of the
technology can be understood.

The ‗communities of promise‘ concept draws on the work of Benedict Anderson.


Anderson‘s (1983) study of the rise of nationalism and national identity describes the
development of nations as ‗imagined communities‘. He describes the development of
nationalism as the outcome of shared imaginations. These allow members of a community
to identify with each other, despite never meeting, and to exclude or ‗other‘ those who do
not share their national imagination. In terms of the technological expectations, imagined
futures enable a shared sense of community and purpose to develop among otherwise
socially or geographically distant groups (Martin, Brown and Kraft 2008).

That nations are ‗imagined‘ evidently does not mean that they are unreal or false, rather that
they are based on the development of a shared sense of identity. This cannot be based on
actual encounters with other members of the community due to the distances involved. The
coordination of these actors within a community of promise is mediated by a shared
imagination. Within the sociology of expectations, distance has been conceptualised in

70
terms of the proximity to basic research, as expectations are described as spatially and
temporally situated (Brown and Michael 2003). Biopharming researchers are thus closest,
while publics and users of technologies are most distant.

Anderson‘s work concentrates on the role of shared imaginations in drawing large


populations together into communities. Although Squier (2004) suggests that the
biomedical imaginary is involved in considering how medical issues are articulated and
engaged with across all cultural fields, much of the existing focus on ‗communities of
promise‘ concentrates on the narrow interactions between biomedical science and interested
publics, particularly patient groups8. As such, discussion of communities of promise
parallels studies of the role of publics in the co-production of biomedical knowledge
(Rabeharisoa and Callon 2004). Each concentrates on the activity of small groups of
interested and engaged publics. Yet the ‗red/green divide‘ described in the previous
chapter, notably in Eurobarometer studies, suggests that biomedical biotechnologies
mobilise support among a wide range of publics.

Anderson (1983) suggests that the rise of shared imaginations was in part enabled through
the growth in the use of vernacular languages in print. As such, national communities align
along linguistic boundaries, particularly in Europe. For example, the publication of
specifically Ukrainian grammars and the publication of literature in Ukrainian is described
as ―the decisive stage in the formation of an Ukrainian national consciousness‖ (1983: 72)9.
Expectations and promises about science and technology represent a similar way in which
communities coalesce around particular linguistic forms. As Brown describes, the language
of rationalistic scientific authority is now supplemented ―with a language drawn from
values and aesthetics with its connections to desire, longing, the imagination and
authenticity‖ (Brown 2007:340) .

The promises of technoscience are portrayed in affective language, an emotional


vernacular. As with the ‗popular, vernacular-based nationalisms‘ described by Anderson

8
An exception to this might be Busby and Martin‘s (2006) discussion of the evocation of national identity in
the development of the UK Biobank DNA repository.
9
Although he suggests that such linguistic unification may not be necessary in an age of multilingual
broadcast media (Anderson 1983: 123)
71
(1983:127), the language of authenticity and hope in technological promises forms part of a
particular form of capitalism. It is this circulation of technological promise that I explore in
Chapter Four in the context of biopharming‘s relation to existing communities of promise
around biomedicine, and corollary communities of apprehension around agricultural
biotechnology.

However, central to Anderson‘s description of nationalism is the role of mediation. He


suggests that the ‗nation-building‘ policies of the early 20th century were accompanied by a
―systematic, even Machiavellian, instilling of nationalist ideology‖ through the media and
state outlets. Yet as Hague (2004) suggests, consideration of whose imaginations are
transmitted is largely lacking from the work of Anderson.

Moreover, Anderson describes capitalism and the development of print technology as


central to the formation of imagined communities, particularly the mediating role of
newspapers and print in allowing the sharing of information and disparate experiences and
events among otherwise heterogeneous groupings. This emphasis on the homogenizing and
driving role of print and text suggests a deterministic account of the role of technology in
the development of nationalism. Adopting Anderson‘s description of societal change as
prompted by the advent of print capitalism represents a move away from the insights of the
social construction of technology and actor-network theory that technology and society are
‗co-produced‘ (Jasanoff 2004).

In the context of technological expectations, Brown (2007) suggests that in communities of


promise different participants ‗conspire‘ or ‗collaborate‘ to constitute distributed
authorship. In this distributed, authorless state, questions of power and of the authorship of
imaginations and depictions of the future would appear irrelevant. However, the work of
Edward Said (1995) emphasises the centrality of power in the shaping of the imagination.

2.1.3 Promising Geographies

In their introduction to the sociology of expectations, Brown and Michael (2003:5) called
for ―a detailed examination‖, not only of the ways in which the future is ‗performed as a
temporal representation‘, but also of the ways in which it is ‗colonized as a spatial and
72
temporal locus‘. Yet the majority of work in the sociology of expectations to date has
concentrated on the former. Here the work of Said (1995) on ‗imagined geographies‘ is of
use, not least in light of Brown and Michael‘s (2003) discussion of attempts to ‗colonise‘ of
the future.

Martin, Brown and Kraft (2008) explore the work of Said alongside that of Anderson in
relation to the promise of stem cells, but with a notable emphasis on the latter.
Consequently much remains to be said on the spatial as well as the temporal features of
technological imagination. As described in the introductory chapter, biotechnologies, and
no less biopharming, are explicitly and inescapably spatial projects.

Said‘s work again highlights the importance of imagination in shaping the behaviour of
social actors. In Orientalism (1995/1978) Said describes how literary representations have
shaped Western understandings of ‗Oriental‘ identity and constitute ‗imagined geographies‘
of the Orient which authorised the colonial projects of Western powers. In these depictions,
the imagination functions over and beyond ―positive knowledge‖ (Said 1995:55) of the
historical or geographical circumstances of the Orient. As Massey et al. (1999) describe,
focusing on imagined geographies contributes to understanding how the ability of groups to
lay claim to territories owes much to the authority vested in their stories, descriptions and
organizing views. As such, it emphasises the interconnections between the narratives and
stories described by the sociology of expectations, and the spatial distribution and
application of technoscience.

Said (1995) suggests that imagined geographies form part of the ‗urge to classify‘ described
by Levi-Strauss, of the fundamental need to impose order on the world. Unlike Anderson
(1983), who describes the role of the imagination in the constitution of shared identities,
Said suggests that the purpose of such geographies is to distinguish oneself from others. As
he puts it, a fifth century Athenian would define himself as much as ‗non-barbarian‘ as
‗Athenian‘. In doing so, particular actions are authorised towards those classified as
‗barbarian‘.

The work of Said introduces the geographical narratives that characterise and constitute
imagined communities, and highlights that they reflect and reinforce distinctions and power
73
relations between actors. In the case of technological innovation, Jasanoff (2006) adopts a
similar ‗post-colonial‘ stance to describe developments in biotechnology as part of the
contemporary ‗colonising‘ efforts of globalization. She warns that ―without institutional
innovations, biotechnology as currently governed may increase the power of metropolitan
centers of science and technology in relation to people at the periphery‖ (2006:277).
Although she does not describe it as such, she identifies what we might term, following
Said, an ‗imagined geography‘ of crisis in the rhetoric of scientific and industrial
development institutions. Within this imaginary ―Africa is represented through tropes of
crisis and charity that render the continent‘s condition as dire‖ (2006: 289). These
representations authorise narratives of salvation through scientific and technological
solutions such as Golden Rice and genetically modified agriculture, together with neo-
liberal practices of governance.

In Jasanoff‘s discussion of biotechnology, the promise of salvation is based on the transfer


of technology from the developed to the developing world, a process itself based on
assumptions that the social and ecological circumstances at the periphery can be, or are,
standardised in the same way as the crops themselves (Jasanoff 2006). Imagined
geographies are, like futures, performative, as they reshape agricultural production in the
image of the envisioned production systems embedded in their development.

As Martin, Brown and Kraft (2008) point out, imagined geographies are not only spatial,
but temporal. They rely on the characterization of imagined futures, but also on depictions
of the present and the past. However, the ‗placing‘ of new technologies in the present and
as part of historical narratives itself involves a work of imagination beyond ‗positive
knowledge‘ (Said 1995).

2.1.4 From Imagined Futures to Authorising Pasts

Both the geographies of Said and the communities of Anderson emphasise the importance
of representations of the past to the imagination. Not only the futures, but the pasts of
biotechnology are challenged as the relationship between biotechnological revolution and
evolution is worked through. However, research considering the role of past narratives has

74
predominantly focussed on bygone visions of the future, or on ‗retrospecting prospects‘
(Brown and Michael 2003; Deuten and Rip 2000; Koch 2006). This work describes how
these retrospections are used to make the eventual trajectory taken by a technology‘s
development appear inevitable (Borup et al. 2006). It suggests that this results from a ‗kind
of historical amnesia‘. As Borup et al. put it in a review of the sociology of expectations

―hype is about the future and the new—rarely about the past—so the disjunctive
aspects of technological change are often emphasized and continuities with the past
are erased from promissory memory‖ (Borup et al. 2006:290)

I draw on another strand of Edward Said‘s work, that on beginnings (1975), to suggest that
the performance of both continuities and discontinuities plays an important role in the
development of expectations. Throughout their development, biotechnologies are related to,
and contrasted with, existing agriculture and medicine. The difficulty of stabilising these
relations reveals the work that goes into the historiography of new technologies. As are
potential futures, salient pasts are not pre-determined, but are defined through narration
(Brown, Rappert, and Webster 2000). Imaginations of the futures and geographies of
technological development thus rely on authorising narratives of technological pasts which
are themselves imagined, constructed and performative.

What Jasanoff (2005) describes as the ‗framing‘ of biotechnology establishes a link


between the hopeful futures described by the sociology of expectations, and wider social
narratives of technoscience‘s pasts and promises. Together, these form an essential part of
placing technologies such as biopharming. Retrospective narrative of the development of
technologies represents a ‗mode of ordering‘ through which we make sense of our present
circumstances (Law 1994:52) and silences the complexity and contingency of actual events
(Deuten and Rip 2000).

Brown (2000) describes how narratives of medical research are typified by iconic events
which act as ‗commemorative moments‘ of success (2000). The ‗breakthroughs‘ and
‗miracles‘ of biomedicine link temporally distant events into a narrative of progress,
lending credibility to hope and creating a moral imperative to pursue future repetition of the
success of these achievements (Brown 1998). Consequently, rhetorics of hope are

75
supported through placing novel technologies within stories which include the past
successes of other technologies. Past heroic or miraculous examples of biomedical success,
often personalised by their discoverer - Fleming and penicillin, Pasteur and anthrax, Jenner
and smallpox –are used to reinforce the claims for novel technologies. While
technologically distinct, they are united by a shared discourse of medical hope, and a
master narrative of technoscientific progress. This is not new - in his manifesto for post-
World War Two funding of scientific research, Vannevar Bush (1945) put forward an
argument based on the success of penicillin.

The discovery of penicillin and other well known events in the history of medical research
serve to support future research and validate forms of governance and funding. Brown
(1998) highlights the 'promotional aspect' of the breakthrough, in terms of directing the
gaze towards the future repetition of past success. Brown‘s accounts of breakthrough
(Brown 1998, 2000) highlight how "breakthrough discourse designates salient historical
nodes and implies the narrative disjunctures of which progress stories are comprised"
(Brown 1998:84).

The construction of analogous relationships between present and past events is intrinsic to
the production and circulation of narratives of future success. Equally though, such
relationships can be used to contest and undermine these narratives, replacing the hopeful,
utopian claims of technological proponents with dystopian visions of ecological and
technological disaster. For example, in UK debates around embryo research, Mulkay
describes how fearful visions of the future were distanced from science fiction and rendered
‗realistic‘ through association with the historical figure of Nazi scientist Dr Mengele
(Mulkay 1993). Mengele anchored opponents‘ interpretations of embryo research in
historical events.

The attribution of what Said terms ‗beginnings‘ in this way establishes the present on a
narrative path – for Said (1975) they provide ‗a place in time‘. As in attempts to link
biotechnology to Babylonian brewing, beginnings are orienting, they establish the position
of technologies ―in a narrative of which the lines and lessons are recognisable and

76
interpretable for the audience‖ (Van Lente 2000:46). In his analysis of beginnings in
literature Said describes their narrative function:

"the necessary creation of authority for a beginning is also reflected in the act of
achieving discontinuity and transfer: while in this act a clear break with the past is
discernable, it must also connect the new direction not so much with a wholly unique
venture but with the established authority of a parallel venture" (Said 1975:33)

Similarly, as Brown describes "the events which come to count ... simultaneously perform
continuities and discontinuities" (1998:72). Said extends his discussion from literature to
attempts to establish new sciences, drawing on the example of psychoanalysis to describe
how:

"the beginning of a new science gains some of its authority when it points toward -
intends - a continuity...Psychoanalysis redeploys old elements, arranging them
discontinuously with, yet parallel to, the traditional manner" (Said 1975: 33)

A novel beginning is thus a negotiation between the continuous and the discontinuous, and
the establishment of continuity is an achievement of skilled narrative positioning. For
biopharming, as for psychoanalysis or xenotransplantation, narrative linking with the
authority of parallel ventures, especially that of biomedical research and pharmaceutical
biotechnology, will be of importance. These narrative links represent a form of association,
which allow comparisons and similarities to be drawn between new and old technologies.
These forms of narration and association contribute to the formation of ‗communities of
promise‘ around novel biotechnologies.

Brown and Michael‘s (2001) discussion of xenotransplantation (XT) highlights that


relevant continuities for novel technoscience are not only those of biomedical research, but
those of more mundane narratives. They examine how experts ‗switch‘ between scientific
and moral discourses in the promotion of xenotransplantation. In the ‗cultural‘ debate
around the choice of donor species justifications for the choice of pigs rely on association
with networks within which pig bodies are already implicated, notably that of food. This
association with prior precedence, the ―we‘ve been doing it for years‖ argument, allows
animal advocates and their arguments to be bypassed as they are differentiated from those

77
publics able to rationally follow the genealogical narrative (Brown and Michael 2001).
Here the origin stories of biotechnology spill over from expert discussion to valorise
particular forms of argument and rationality in public debate. I explore the relation between
biotechnology‘s ‗beginnings‘, its futures and its publics in Chapter Four.

However, there are limitations to discussing biopharming solely in terms of narrative and
discourse. Jasanoff (2005) suggests that frames are embedded in the material form of
technologies, yet the focus on discourse and rhetoric of frame based approaches makes it
difficult to effectively foreground these materialities. Possible futures become embedded in
the materials of novel technoscience, and it is through these materials that it is encountered.
As Brown and Michael describe, ―the discursive shaping of the future highlights limitations
when practical and material considerations fail to play along‖ (2003:7). In their work on
classification, Bowker and Star describe how telling stories, both prospective and
retrospective, requires ―objects in the world that can be cut up spatially‖ (Bowker and Star
2000).

2.2 Making Expectations Matter

The ‗biomedical imaginary‘ and breakthroughs of past medical research provide powerful
support to the hopeful futures of biotechnology. However, as Michael (2000) describes, the
performativity of expectations

―is conducted in material settings, where bodies and texts, for example, come into
contact or close proximity‖ (Michael 2000:33)

The products and artefacts of technoscience are central to the performance of expectations.
Indeed, challenges to the development of both green and red biotechnologies have emerged
as their products took their place among those of existing foods and pharmaceuticals. To
expand on this discussion, I describe how both experts and publics relate novel food
products, including first generation agricultural biotechnologies, to their existing
classifications of food. While narration and framing represents one means through which
this takes place, work in STS has described how these imaginations and visions are

78
‗inscribed‘ in the technical content of the artefacts of novel technoscience (Akrich 1992;
Brown and Michael 2003). As Brown, Rappert and Webster introduce:

―Like all discourses, ‗the future‘ is constituted through an unstable field of language,
practice and materiality in which various disciplines, capacities and actors compete
for the right to represent near and far term developments‖ (2000: 5; my emphasis)

The materials of novel technologies both embody their promise and ‗pre-discipline‘ the
imagination (Brown 2007). The products of new biotechnologies also provide the primary
site at which these technologies are encountered by regulators, publics and investors. As
Borup et al. put it, the artefacts of technoscience are ―materialized indicators of
expectations‖ (2006: 292). It is through these processes, the embedding of expectations in
new technologies and their reception, that the novel ontologies of biotechnology can be
classified and ordered. For Brown and colleagues, this enables the sociology of
expectations to be ‗a symmetrical sociology of the future‘ that explores the connections and
dissonances between the materiality of technoscience and its articulation in language
(Brown et al. 2000:15).

This form of ‗placing‘ or ordering of biopharming and its expectations echoes the
traditional concerns of STS around questions of categorisations, purifications, boundaries
and associations (Gieryn 1983; Bowker and Star 2000; Latour 2005). I draw on discussions
of ‗relational materiality‘ and performativity (Law 2002; Latour 2005) which describe how
entities take and maintain their form and acquire their attributes as a result of position in
relation to others.

In particular, I am interested in exploring the relationship between the novel products of


biopharming and the mundane objects of everyday foods and medicine. The artefacts of
technoscience ―change their ‗identity‘ by virtue of the associations they form in the process
of ordering and disordering‖ the patterns of everyday life (Michael 2006:155). Michael
goes on to suggest that technoscience and everyday life are interwoven and heterogeneous,
and the former is ‗located‘ within the latter. Among the other locations described in
Chapter One, he suggests technoscience is ‗located‘ when it is:

79
―mundanely manifested in the practical and often unnoticed technologies (and
expertises) that cohabit the everyday with us: refrigerators, computers, cookers,
radios, televisions, washing machines, beds shoes, tables, armchairs, clothes,
mirrors, envelopes, pens, paper, light bulbs, cars, trains, knives, zips, velcro, ring
pulls … here the list tumbles out of control. I include these as parts of technoscience
because they were, once, in one way or another, before they became more or less
invisible technologies, inventions‖ (Michael 2006:33)

Michael‘s tumbling list of mundane items illustrates that today‘s novel technoscience
becomes tomorrow‘s polystyrene chip box. It suggests that the knowledges that locate
technoscience within the everyday are broader than the scientific. Michael continues to
describe how ―regular intrusions of the novel and the exotic‖ are ―intrinsic to the modern
everyday‖ (Michael 2006: 28).

These ‗intrusions‘ of the exotic into the everyday reveal conflicting ontologies of personal,
natural and moral order (Davies 2006) as a key site of encounter between experts and lay
actors at which each attempts to place the novel among their conceptions and expectations
of the existing. As described by Jasanoff (2005), novel agricultural and food
biotechnologies must make a place for themselves in competition with the products of
existing agriculture. Work within what Latour (2005) describes as the ‗sociology of
associations‘ suggests that the attributes and forms of these novel biotechnologies are also
produced through this placing. In my empirical discussion, I will explore how biopharming
similarly engages and is transformed through with the everyday artefacts of existing
agriculture and medicines. In introducing this approach, the relationship of GM crops to
existing foods provides a useful example. It highlights how the relationship between GM
and agriculture sits within and is co-produced with wider challenges to food production,
and consumer engagement with these.

The relevance to biopharming of encounters between novel applications of agricultural


biotechnology and everyday foods can be highlighted through Jasanoff‘s (2006) discussion
of ‗golden rice‘, a ‗second generation‘ agricultural biotechnology with similarities to both
the technology and promise of biopharming. The categorisation of golden rice seems
straightforward, particularly for proponents who claim it is nothing more than a more

80
nutritious plant variety. However, as Jasanoff describes ―food crops straddle too many
categorical boundaries for their identity in the political domain to be anything but hybrid‖
(2006:287). The relationship between golden rice and food is not simple, as food crops are
not themselves easily delineated.

I exemplify this point in terms of substantial equivalence and ‗comparative analysis‘, which
establish existing food products as the relevant reference point for novel biotechnologies. I
then examine how this relational establishment of food qualities parallels the discussion of
consumers around the qualities of novel and existing foods. I consider how expert and lay
contexts for the introduction of novel biotechnologies are co-constructed in relation to
everyday foods.

The examples presented here illustrate how bringing together expert and lay perspectives
reveals the respective producer and consumer roles assumed by experts and publics of
biotechnology. I draw further on the work of Brown and Michael (2001) introduced in the
previous section to consider how repertoires through which the novel ontologies are related
to existing products are accepted or rejected as they are interpreted by expert and lay actors.

2.2.1 The proof of the pudding is … in structural and functional similarity?

Burchell (2007) identifies a ‗discourse of similarity‘ among experts which asserts that GM
techniques are part of a continuum with conventional modern plant breeding techniques.
This represents the narrative placing of GM, but also establishes relations between the
matters of agricultural biotechnology and those of existing food production. The product
approach to biotechnology regulation described by Jasanoff (2005) concentrates on ―the
structural and functional similarity of GM and ‗natural‘ products‖ (Jasanoff 2005:133).
Attempts to base regulation of biotechnology on its products posit a similarity between the
products of biotechnology and those of other production methods. To be approved, a novel
food must be at least as safe as the conventional food that it will replace.

However, Millstone et al. (1999) criticise this approach on the basis that as well as not
incorporating sufficient criteria, it does not define the point at which substantial
equivalence ceases – there is no juncture at which dissociation between GM and
81
conventional crops can occur. In Canada, Barrett and Abergel similarly describe how
reliance on substantial equivalence ―forecloses questions of uncertainty, marginalises
sceptical and critical voices, and effectively excludes the public from risk debates‖
(2000:3). As a means of determining how associations are made, it has consequences for
the subsequent development of the technology and its relations to broader contexts of food
production and the democratic process.

As Levidow et al. (2007) chart, criticisms of substantial equivalence were partly recognised
in EU Regulation 1829/2003, in which substantial equivalence evolved into a ‘comparative‘
approach to the assessment of GM foods involving assessment of composition, toxicology
and allergenicity in a more open-ended policy framework. This approach was outlined by
EFSA GMO scientific advisory panel in 2006 as a two step procedure:

―The first step ... is the comparative analysis of the molecular, agronomic and
morphological characteristics of the organisms in question, as well as their chemical
composition.. The outcome of this comparative analysis will further structure the
second part of the assessment procedure, which may include further specific safety
and nutritional testing‖ (EFSA 2006:13)

The bases on which association is made have shifted and substantial equivalence is now
‗downgraded‘ to the first step in the regulatory process (Ely 2008). The analysis involves
the generation of new knowledge about the specific novel characteristics of the plant, but
regards the more general features of foods as familiar. The development of substantial
equivalence regulation demonstrates the heterogeneous scientific, regulatory and social
relationships which go into constituting GM crops as foods.

Thus, as in discussion of expectations, debates around substantial equivalence highlight that


the identity of biotechnologies is emergent, a process of negotiation and stabilization. These
identities are formed as the ‗intrusion‘ of the products of novel technoscience into the
contexts of everyday life is experienced and contested by publics and regulators. In the
following section, I extend this discussion to examine how publics similarly place the novel
within the existing, and highlight the parallel knowledges to those used by experts that are
drawn on in delineating and discriminating between the products of food and medicine.

82
2.2.2 The proof of the pudding is … in looking and touching?

The example of substantial equivalence suggests that biotechnology prompts ―a new


analytic engagement with the materiality of nature…as an ontologically real and active,
lively presence‖ (Goodman 2001:183) for both experts and diverse publics. As
demonstrated in recent research, the public practices of quotidian food consumption play a
vital role in shaping the complexity of these ‗food landscapes‘ (Roe 2006).

First generation genetically modified crops emerged into a highly visible societal
discussion in Europe and North America about food (see for example Lawrence 2004;
Pollan 2007). This frames publics as active and discerning consumers, both able and duty-
bound to judge and influence the production and quality of food. In understanding how
novel biotechnologies are placed among foodstuffs which are themselves hybrid and
contested it is thus necessary to follow Roe in asking ―what skills and knowledges are
applied to construct the edibility of a foodstuff for the contemporary consumer‖ (2006:
472)?

Discussion of how publics-as-consumers think about and distinguish foods-as-commodities


emphasises the importance of knowledge derived from their aesthetic qualities and
appearance (Green et al. 2003; Eden et al. 2008). Eden and colleagues draw on work in
consumer studies to describe how a range of qualities contribute to sorting ‗good‘ from
‗bad‘ foods. In particular, visual assessment of foods provides direct empirical evidence of
their qualities, both in terms of health and in relation to their mode of production. Green et
al. conclude that in the absence of such direct empirical evidence foods are adjudged to be
less safe and more ‗risky‘ (Green et al. 2003:47).

The role of the displayed qualities of foods in this work is in the production of knowledge
about their intrinsic or invisible qualities: whether they are healthy, safe and how they have
been produced. In the absence of ‗primary characteristics‘ such as appearance and taste,
Eden et al. (2008) describe the presence of ‗proxies‘, such as where food is purchased and
reliance on labelling and quality assurance schemes. Following Latour (1988) they describe
how quality assurance ‗black boxes‘ food growing and processing, ―providing a
standardised shorthand…for quick purchase decisions‖ (2008: 1054). Similarly, Green et
83
al. (2003) describe the ‗rules of thumb‘ that consumers use as ‗shortcuts to safety‘ - ―simple
dualities that divided the `good' from the `bad' in terms of food choices‖ (2003:39). These
include distinctions between organic and non-organic; ready made and home made;
traditional and mass-produced.

Much of the research introduced heretofore emphasises the importance of the visual
characteristics of foods in relating them to typologies of quality and safety. However,
further work in agro-food geographies reintroduces the materialities of food and the
ontologically real and lively presence described by Goodman (2001). Stassart and
Whatmore argue that the traditional focus on production within agro-food geographies has
led to a neglect of food itself and its materiality (Stassart and Whatmore 2003:450). Roe
(2006) builds on this critique to consider how the meanings attributed to foodstuffs become
contingent. Using the example of carrots, she explores the relationship between genetically
modified, organic and processed varieties to describe a process of ‗becoming food‘. The
work of Roe (2006) and Eden et al. (2008) emphasises that the meanings of food qualities,
and the expectations they embody, are not stable. For example, rather than establishing
quality through the removal of dirt, in ‗alternative‘ food networks, dirt and mud is an
important ‗primary characteristic‘ often intrinsic to the authenticity of food products.

Roe (2006) brings together the cognitive ways of knowing described above with corporeal
experiences of eating to understand how we come to `know' what to eat and what not to eat.
She points to the importance of three levels at which materiality is a concern, in the form of
integral, superficial, and what she describes as ‗immaterial‘ materialities. Roe pursues the
materiality of food through focus group studies of how food becomes edible or inedible,
how a ‗thing‘ comes to be ‗a food‘. In attending to the ways in which participants in her
research talk about the way they consume and prepare food, she examines the material
‗connections‘ which constitute edibility. She examines how edibility is performed through
not only cognitive, but affective and the social connections. In moving on to discussion of
biopharming, the forms of ‗material connectivity‘ and the different ways in which it is
performed described here will be useful in thinking through how its products are placed in
relation to existing food and medical products.

84
In considering conventional foodstuffs, consumers construct their understandings of
edibility through more than appearance. As Roe puts it:

―The plants and animals that become our food are tested through our powers of
smell, touch, taste and sight and it is only after passing these tests does the food,
through digestion, become integrated into our bodies‖ (2006: 118).

Moreover, as Grunert (2002) and Eden et al. (2008) highlight, the material qualities that
provide a first set of cues about foods quality are accompanied by a range of secondary,
characteristics which act as ‗proxies‘. Consequently, expectations of food quality are
derived from information in packaging and labelling. For example, the suggestion of
labelling for genetically modified foods relies on the consequent re-assertion of consumer
choice (Einsiedel 2002). In addition, as I explore in section 2.3, the qualities of foods are
also established on the basis of ‗spatial proxies‘, such as the locations of food production
and consumption.

As reproduced in figure 2.1, Eden et al. (2008) provide a typology of knowledge topologies
which describes the relationship between producer and consumer, something that may be
useful in comparing the production of public knowledges about food commodities with
those of pharmaceuticals. This typology emphasises the existence of multiple different
networks of food consumption, and the role of heterogeneous intermediaries, entities and
places. Most interesting in the current context is the role of assurance and verification in
enabling the ‗black-boxing‘ of food production. In the case of food scares such as BSE or
GM, these black boxes are opened and the meanings attached to foods shift as consumer
knowledges and practices contest those of 'experts' (Stassart and Whatmore 2003; Jasanoff
2005). As I will explore in Chapter Five, the role of intermediaries in the provision of
information provides a starting point to consider contrasts between the roles adopted by
publics in relation to food and pharmaceutical products.

85
Figure 2.1 Simplified knowledge topologies and black-boxes for different
types of food consumption. From Eden et al. (2008)

2.2.3 The importance of association and the problems of consumption

The consequences of relating the products of novel technoscience to those of everyday life
are highlighted in the case of xenotransplantation described by Nik Brown and Mike
Michael (Brown and Michael 2001; Michael and Brown 2004). The work of Roe (2006)
introduces the contingency of meanings attributed to foods. Brown and Michael (2001;
Michael and Brown 2004) describe how the meanings attached to the materialities of
xenotransplantation became contingent as expert and lay understandings of meat conflict.
Relating the novel to the existing normalises it within familiar narratives. However, this is
reliant on aligning the qualities of the new products with existing materials.

Brown and Michael (2001) describe how scientists ‗switch‘ between the cultural repertoire
that links traditions of meat eating and the use of pig organs in XT, and a scientific one

86
which argues the physiological similarity of porcine and human bodies. This discourse
presents relations of sameness and difference between species as direct statements of fact
about ――rational‖ and ―objective‖ ratios of size, physiological parity, and so on that serve as
the grounds for similarity between prospective donor and host‖ (2001: 12).

Following from this earlier work, Michael and Brown (2004) examine the statement that
―there is no difference between using pigs for xenotransplantation (XT) and eating a ham
sandwich‖. This analogical assertion positions XT alongside existing practices and
everyday products. Moreover, the audience of this statement is being asked not to compare
XT to meat eating, but to understand XT in terms of meat. A direct association is being
made between the novel materials of biotechnology and those with which people are
familiar.

Arguing for equivalence between XT and meat serves as a means of countering the disgust
or ‗yuk factor‘ of the material mingling of animal and human bodies by normalizing it,
making it no longer ‗matter out of place‘ (Burke 1998; Midgley 2000; Douglas 2002). As
such, it may be important in approaching biopharming. However, as Michael and Brown
(2004) suggest, the complex meanings attached to meat make associations with it
problematic as expert and lay understandings collide. The use of the meat analogy does not
uncomplicatedly equate XT with the naturalness and purity of meat, as contemporary, post-
BSE meat is itself often understood to be impure and unnatural. Moreover, although
Michael and Brown do not explore this, the analogy also implies particular, carnivorous,
consumers. Michael and Brown (2004) argue that the analogy therefore only works in some
framings, notably as experts equate XT and meat on a functional and production basis. The
analogy succeeds along certain dimensions, such as ―functionality‖, in that as ―meat
enables the functioning of the body, so too does the xenotransplanted organ‖ (2004:393).
Equally, the use of animals as a production vehicle establishes an equivalence between the
two processes.

However, discussing the relation between eating and transplantation, and focussing not
only on production and function, but on the ―process and unit of ‗incorporation‘‖ (2004:
393), acts to dissociate XT and meat. One way of portraying this distinction is as between

87
expert ‗producers‘ and lay ‗consumers‘, the former who understand food in terms of
production, the latter in terms of consumption. However, both ‗producers‘ and ‗consumers‘
are heterogeneous categories.

While discussion of meat consumption challenges the association with xenotransplanation,


a narrowly scientific repertoire which asserts similarity between pig and human bodies
protects this from similar contestation. As Brown and Michael (2001) put it, the relations of
difference and similarity inscribed in the identities of donor bodies extends into the
identities of experts and publics. They suggest that ―the recourse to descriptions of the
physical properties of animals‖ demarcates expert domains (2001: 13). Only experts are
able to speak on the immaterial and unspoken characteristics of things.

The example of XT suggests that the qualities of novel products such as those of
biopharming are known through metaphor and analogy with existing ones. However,
Michael and Brown (2004) suggest that parallels drawn between an innovation and a
familiar object may lead neither to ease of understanding nor to confidence in judgment.
Not all analogous relationships are equally applicable or stable – they are politically
freighted and the source of the analogy may not be trusted. Consequently they vary in their
persuasiveness.

The failure of analogous reasoning between XT and meat to establish the promise of the
technology fails not because of deficiencies in the argument, or necessarily because of the
nebulous nature of XT, but because of the shifting and contingent meanings of meat itself
as the object of consumption. Similarly, in the case of biopharming, encounters between
expert and lay understandings of the material qualities of food and medicines will
contribute to placing it in relation to foods and medicines.

2.2.4 Between Consumers and Patients?

In the case of xenotransplantation, the positioning of xenotransplantation in relation to meat


is introduced by experts, as part of attempts to normalise the harvesting of human organs
from pigs. However, the meanings they attach to the materials of XT are unstable due to the

88
complex meanings attached to meat by publics. This establishes a divide between experts as
producers and publics as consumers.

Since the 1980s, consumption has become an integral part of the expression of citizenship,
as a means of expressing desires and concerns beyond those narrowly limited to the
purchasing of food. Michael (1998) and Irwin and Michael (2003) elaborate on the role of
consumers in terms of the public understanding of science. They suggest that the identity of
the ‗consumer‘ has become a central one through which citizenly identities and democratic
expression are performed, including around novel technoscience. While Irwin and Michael
(2003) highlight that the relationship between citizen and consumer identities can be
articulated in multiple ways, in the case of genetically modified crops food choice played a
prominent role (Frewer and Salter 2002) and undoubtedly spurred corporate attempts to
engage with the concerns of consumer-citizens (Doubleday 2004).

As Doubleday (2004) describes, the empowerment of consumers in debate around


genetically modified crops lead to the performance of citizen-like behaviour in relation to
private companies as well as nation-states. However, in considering biopharming, the
extent to which such consumer-citizens can be observed in considering pharmaceuticals is
unclear. In the context of expectations, Martin, Brown and Turner (2008) suggest that

―links between markets and promissory expectations depend on the mobilization of


publics as active consumers within newly neo-liberalized bioeconomies‖ (Martin,
Brown and Turner 2008:129)

Clearly, in the ‗communities of promise‘ described in section 2.1, the role of publics is one
of active and informed consumers similar to those described in the context of foods. While
a number of studies examine the performance of this consumer role among small groups of
patients or interested groups such as ‗prudently responsible parents‘ (Martin, Brown and
Turner 2008) there are fewer studies that focus on publics as everyday consumers of
medicine.

The quotation from Martin and colleagues suggests that biopharming is emerging in the
context of wider changes in the delivery and development of novel pharmaceuticals. The
recent development of medicine has seen moves to a ‗consumer health system‘ in the UK
89
(Pollock 2004) and from ‗patient‘ to ‗consumer‘ identities more widely (Applbaum 2006).
Building on the legacy of 1980‘s Thatcherism, a decade of New Labour governments have
continued a shift to public services in which the relevant publics are consumers. As one UK
Health Secretary, Alan Milburn, put it, ―people expect choice and demand quality‖
(Milburn 2001 in Vidler and Clarke 2005), while the ‗Choosing Health‘ White Paper of
2004 points out: "many of the choices that affect our health are choices we make as
consumers" (Department of Health 2004).

Despite this shift, most existing studies in medical sociology have focussed on publics as
consumers of prescription medicines. They emphasise that these consumers do actively
participate in the construction of these prescriptions, and describe how patients may refuse
to comply with their prescriptions (Cohen et al. 2001; Webster, Douglas, and Lewis 2009).
However, their description of mediated access to medicines provides a poor basis on which
to consider the role of the materialities of pharmaceuticals in a way that parallels active
consumer engagements with foods. Stevenson, Leontowitsch, and Duggan (2008) provide
one of few studies that consider the consumption of medicines in similar terms to that of
foods. They highlight the emergence of discussion of pharmaceuticals as commodities, in
which ―pharmacies may be used like any other shop to purchase a particular product‖
(Stevenson et al. 2008:925). Interestingly, the examples they use from public discussion are
those of cough sweets, which may themselves be considered as existing on the borders of
medical products and foods.

However, Stevenson et al. (2008) also suggest that the relationship between consumers and
pharmacists in purchasing ‗over-the-counter‘ medicines is one of an asymmetry of
knowledge, and that most consumption of these medicines involves a relationship between
experts and publics in which expert knowledge is dominant. In terms of the topologies
introduced above, pharmaceutical production most closely approximates that of
independently certified production, in which the entire process is black-boxed.

Contemporary health provision emphasises the role of publics as consumers of health


products. Consequently, medical sociology is increasingly focussing on users of
pharmaceuticals as knowledgeable actors (Williams, Gabe, and Davis 2008; Webster et al.

90
2009). Yet Stevenson et al.‘s (2008) paper represents one of very few examples of how
publics as consumers or patients exercise choice and discriminate between pharmaceutical
products. As Ecks points out in a commentary on Hayden‘s (2007) ethnography of generic
medicine provision in Mexico:

―The single most important characteristic of pharmaceutical markets is that nearly


everyone who spends money on drugs has no idea about who was involved in their
production and distribution, who is making profits, or how quality is secured. This
applies just as much to generics as to brand medicines. All that is ―public‖ … is the
active ingredient‖ (Ecks in Hayden 2007:490)

According to Ecks, regardless of whether pharmaceutical provision is state or market led,


there is little public awareness of their production, quality assurance or of corporate control.
The work of Abraham and Lewis (2002) would suggest that this is due to the lack of
availability of similar information to that which characterises consumer discussion of foods.

Abraham and Lewis (2002) describe the emergence of an ‗active citizenship‘ among
pharmaceutical consumers from the 1970s onwards, particularly following the catastrophic
failure of regulation in the case of thalidomide. Here parallels might be seen with the rise in
consumer engagement and knowledge, and decline in confidence in food safety following
the multiple scares of the 1980s and 1990s, culminating in BSE (Stassart and Whatmore
2003). However, active public engagement with pharmaceutical biotechnology has been
minimal. Moreover, Abraham and Lewis (2002) suggest that although consumerist
expressions of citizenship may have become more common, in the case of pharmaceuticals
there has been little or no erosion of producer authority. Hence the information about the
safety and efficacy of medicines required to make consumer decisions and to ensure the
transparency of the pharmaceutical industry has remained subject to intellectual property
law. Attempts to release this information, such as the 1993 Medicines Information Bill,
have been thwarted by agreement between the British government and the pharmaceutical
industry, such that ―manufacturers‘ rights to private property were privileged over
consumers‘ rights to security in health‖ (Abraham and Lewis 2002:74).

Given the relative paucity of consumer discussion of pharmaceuticals, in considering how


the products of biopharming relate to those of existing production methods, in examining
91
how the products of biopharming are placed by publics it may be instructive to draw on
discussion of foods, where the identification of publics as consumers is perhaps more
deeply entrenched.

The approaches described here point to the need to combine analysis of symbolic and
material associations between biopharming and existing biotechnologies, foods and
medicines. The juxtaposition of public and expert discourses highlights the existence of
parallel practices of association and differentiation. The work of Green et al. (2003), Eden
et al. (2008) and Roe (2006) highlights the importance of a range of characteristics of foods
in relating them to wider concerns, including their safety, but by no means limited to this.
They highlight how a focus on the ‗risk‘ of food consumption ignores the role of quality, of
health considerations, of ethical consumption practices, or of the valorisation of particular
forms of agricultural production. In contrast, the practices described in the context of
substantial equivalence reflect a narrow expert focus on safety as the defining
characteristics of food.

For both lay and experts actors however, the qualities of the products of technoscience,
both exotic biotechnologies and the mundane technologies of farming, are established in
relation to other existing products, institutions and actors. These products carry and embody
expectations of particular outcomes, and are the means through which these expectations
are encountered and challenged. As such the relational establishment of the qualities of the
products of food and technoscience will be important for considering how the promise of
biopharming is approached by expert and lay actors.

2.3 A Place for Everything, and Everything in its Place

As Anderson et al. (2007) describe, technological change is typically represented in terms


of the development of new techniques ‗in time‘, rather than in space. As my discussion of
‗imagined geographies‘ describes, spatial and temporal futures are not easily separated.
Moreover, the practices of material association described in the previous section are
accompanied by material geographies of science, food and pharmaceuticals.

92
Of the definitions of place introduced by Staeheli (2003), existing work on technological
expectations has primarily used a concept of 'social location'. Brown (2007) describes how
expectations have both a 'temporal patterning' and a 'spatial geography', suggesting that:

―Expectations … are ―situated‖ inasmuch as, for example, bench scientists might be
more jaded or even disillusioned by the sheer difficulties they face in drawing that
promissory imagined future into the present. These uncertainties become less visible
the further one travels in space and time from the material messiness of the
laboratory. Policymakers, investors, even sociologists of expectation are rarely privy
to the private reservations of the laboratory and the biotech boardroom‖ (2007: 336;
my emphasis)

Expectations are ‗situated‘ – they are partial, located and embodied, related to actors‘ social
location. For Brown and Brown and Michael (2003), the geographies of expectations are
constructed through relations of social and temporal distance. Similarly, Borup et al. (2006)
limit their discussion to the 'socio-spatial' patterning of technological promise. Given this
focus on social location and actors, the geographies described by this existing work are
those of ‗expecting‘ – of the formation and circulation of promises, rather than the content
of the visions themselves. As the quote from Brown suggests, the consequences of these
geographies relate to the ability of actors to critically assess the content of expectations.
Elsewhere, Brown and Webster suggest that

"expectations are usually much more optimistic for those who are somewhat distant
from the complexities and uncertainties of innovation and knowledge-making"
(Brown and Webster 2004:181)

In these terms, actors‘ approaches to expectations are thus shaped by the ‗distance‘ from
innovation. Those who are further from the act of promising are likely to see expectations
as having greater authority, and have greater confidence in them. Meanwhile, those who are
making promises are likely to vary the force of their promise depending on their context,
being more cautious in research settings and perhaps more strident in commercial ones.

As Brown and Michael (2003) continue, these distances are relative to networks into which
new technologies are introduced. They suggest that where existing socio-technical
networks are firmly established.

93
―It might … be the case that, for potential users of an innovation, we would see here
a diminished capability to be cautious or uncertain about expectations emanating
from emerging networks and activities. This might be understood as future
‘naivety’, a consequence of distance from the uncertainties of knowledge
production‖ (Brown and Michael 2003: 16; my emphasis)

Uncertainty and scepticism are thus situated in relation to the technology itself. However,
while this extract suggests that a lack of scepticism or caution may be an expression of
‗naivety‘, it leaves the construction of this naivety unexamined, in particular the
distribution of power and knowledge between actors in established and stabilised networks.

However, as well as reflecting the force and distributing of expecting, spatial imaginaries
are often central to the content of expectations, not least in the form of a movement from
‗bench‘ to ‗bedside‘, or from basic research to clinical application (Locating Technoscience
2008; Martin, Brown and Kraft 2008). Spatial imaginaries also serve to support the promise
of technoscience, for example in telemedicine or telework (Geels and Smit 2000; Brown
and Webster 2004). Information technologies often embody utopian visions of the ‗global‘,
in which technology shrinks distance and brings humanity closer together (Bowker 2007).

Although existing work emphasises the situated nature of expectations and suggests
consequences for the assessment of these putative futures, the ‗geographies‘ of expectations
have not been widely considered. The work of Eames et al. (2006) provides an example of
how such research could be approached. Eames et al. (2006) introduce a discussion of
contested normative visions of the future of the use of hydrogen in transport and energy.
They highlight how three visions of the hydrogen economy are performed with reference to
London. In particular they suggest that the interpretive flexibility of London as a place
produces an emphasis on different combinations of the mobilising narrative themes of the
‗hydrogen economy‘, and the elaboration of competing visions.

Although it extends discussion of the role of place in expectations beyond that of social
location, Eames et al.‘s description of visions for London‘s hydrogen economy relegates
their geographies to those of ‗context‘. This thesis contributes to the extension of this
discussion of the geographies contained within representations of technological futures.
Following the approach described in the previous section, I explore both the discursive and
94
rhetorical geographies of promise, and their material expression. To do so, I draw on
studies in STS and in the geographies of science which have pointed to the constitutive role
of the material geographies and places of science. I suggest that biopharming's emergence
as a pharmaceutical technology may both be best understood through a consideration of
how it combines agricultural and biomedical places and quality regimes. I also suggest that
it provides an opportunity to extend discussions of place into the otherwise ‗black-boxed‘
topologies of pharmaceutical production.

2.3.1 Places of Technoscientific Quality

As Henke and Gieryn (2008) describe in their contribution to the STS Handbook, with the
advent of laboratory studies, the sites of science became important as locales within which
knowledge was 'constructed' and stripped of modalities, taking on the placeless identity
previously assumed. For example, early work in actor-network theory was firmly grounded
in local contexts, from Latour's account of Pasteur's success in establishing his laboratory
as an 'obligatory passage point' (Latour 1993) to Callon's oft-cited study of the scallops of
St Brieuc Bay (Callon 1986). Although the locations of both these examples are central to
the science, neither is considered in detail, but rather provides scenery for discussion of the
machinations of actors. However, as Finnegan puts it ―space need not be thought of as a
'container' or 'backdrop' for social life...but rather as an active ingredient in social and
cultural life‖ (2008:371).

Science is not simply ‗spatialised‘, but creates spaces, places and geographies (Naylor
2005). Further studies have concentrated on the sites of science as those of knowledge
production, such as Latour and Woolgar's examination of 'Laboratory Life' (1988) or
Knorr-Cetina's laboratory studies (1999). Here technoscience is considered as a process of
production of reliable knowledge about the world and of novel entities within it. As with
other processes of production it has its places, its ‗factories‘ in which knowledge is
‗manufactured‘ (Knorr-Cetina 1999).

In considering the relevance of this work I am particularly interested in exploring how the
qualities of the products in biopharming relate to, and are warranted by, their places of

95
production. I explore how the boundaries of physical place act to ensure the quality and
‗purity‘ of science and define its relation with the spaces of everyday life. The places of
science involve an image of order and control required for the practice of experimental
science which in turn contributes to the development of the identity of science itself and of
scientists. Moreover, these places act to protect science and to enable control over the social
and material disorder and complexity without their boundaries. I focus on the relationship
between two particular scientific places, the field and the laboratory, in the production of
credible knowledge.

Historical studies of the geographies of science describe the central role of place in the
early establishment of science and the way in which credibility for science was constituted
locally (Ophir and Shapin 1991; Livingstone 2003; Powell 2007). As Shapin describes
―science is undeniably made in specific sites, and it discernibly carries the marks of those
sites of production‖ (1995:306). As science moved into new sites of science, such as the
Cavendish laboratory in Cambridge or Thomson's laboratory in Glasgow, new relations
were configured between science and religion and science and industry. As Livingstone
puts it, ―the physical laboratory emerged as the spatial articulation of a new cultural order‖
(2003: 27). While the majority of this work focuses on the historical geographies of science
(Powell 2007), Wainwright and Williams (2008) have recently begun to apply this research
to contemporary laboratory studies, suggesting that similar conclusions can be drawn. They
argue that the places of stem cell research shape and are shaped by the development of
research, describing a series of places of ‗production‘ and ‗acceptance‘ of science.
Following Wainwright and William‘s (2008) example, I explore how the places of
technoscience contribute to understanding how expectations and relations are established
for biopharming, biomedicine and agriculture.

Shapin (1988) describes the inaugural role of place in the development of modern science
and the assertion of its quality, particularly the house on Pall Mall, London, in which
Robert Boyle conducted his experiments with the air pump. For Boyle's experimental
findings to be accepted as 'matters of fact', they had to be attested by people authorised to
testify to the veracity of his claims. In 17th century England, the production of knowledge
was inseparable from norms of conduct between gentlemen and the link between the
96
identities of a witness to science and a gentleman guest in the house of another. Gentlemen
could be relied upon to attest faithfully to the results of experiments, removing the need to
repeat the results in different locations. It is this witnessing that solves the problem of trust
in science, asserts its quality and allows locally produced knowledge to become global.
Kohler (2003) suggests that this remains the case, describing the scientific laboratory as the
socially homogeneous descendant of Boyle's experimental space. Just as in 17th century
England, access is restricted to those qualified to witness. From ―gentleman in, genuine
knowledge out‖, Kohler (2003) suggests that it is now ―white coat in, genuine knowledge
out‖.

The laboratory places of knowledge production guarantee the perceived authenticity and
reliability of scientific knowledge claims through the ways they combine public and private
space and regulate access to production processes. As Gieryn puts it:

―On one hand, experimental laboratories are open places where phenomena, cells,
quarks, intelligence are rendered visible, using procedures and instruments available
in principle to inspection by any skeptic. On the other, laboratories are excluding
spaces, designed precisely to control or discipline phenomena by protecting them
from potentially destructive intrusions of natural pollutants or social
threats.‖(2002:48)

Laboratories act to both include and exclude, controlling those people and entities with
access to knowledge production. They contain the fabrication of knowledge, and by doing
so, contain its social and political encounters. In contrast to the controlled constituency and
geography of the laboratory, the development of field science is characterised by the
diversity and heterogeneity of its participants.

Kohler (2003) argues that the border between laboratory and field science is one of the
most important in the cultural geography of modern science, demarcating standing and
credibility, location and forms of scientific practice. Livingstone (2003) describes the
difficulties of establishing field science as of equivalent quality to that emerging from
enclosed places of knowledge production. He points out that unlike other sites of scientific
practice such as the laboratory, the field is less easily defined, bounded and policed;
inhabited by a shifting cast of actors among whom social relations are defined in the same
97
way as in laboratories, but in a more analytically apparent fashion. Consequently examining
science in the field reveals the ‗formal and informal disciplines‘ which enable stability to be
achieved (Livingstone 2003).

However, as the example of Losey et al.‘s work on the Monarch butterfly (see Chapter
One) suggests, the relationship between laboratory and field is unstable and constantly
renegotiated as scientists attempt to demarcate the boundaries between sites of science and
those of non-science (cf. Gieryn 1983). Kohler (2003) describes how, in the late 19th
century, the failure of evolutionary morphology, the attempt to describe the stages of
evolution from the development of marine embryos, led to recognition that biological
laboratories had become too 'unnatural' and placeless. The failure of the research
delegitimised the places of science in the same way that the early success of researchers
such as Boyle legitimised them. Kohler quotes the comment of one observer that ―the idea
of learning biology proper in a laboratory or a museum is as preposterous as the idea of
learning navigation from a toy ship on a mill pond‖ (Ritter, in Kohler, 2003:29).

In response to the criticisms of laboratory biology, a series of attempts to 'renaturalise' the


laboratory were undertaken to improve the quality of research. Laboratories were situated
in regions similar to those of the climate they simulated, to more closely approximate the
natural, such that plants and animals could be examined in conditions that were controlled
but also natural. As Kohler (2003) puts it, labs ‗merged into nature‘ to improve the quality
of knowledge about the natural world produced at each site.

While Kohler suggests they ultimately met with little success, attempts to ‗renaturalise‘ the
lab emphasise how the placing of science acts to demarcate and define the relationship
between natural and the scientific space such that only sufficient nature is admitted to
ensure the production of quality scientific knowledge. Biological farms and vivaria
underscore the movable boundary between the laboratory and the field, or rather they
suggest that some places of science are hybrid, displaying features of both types of places.

As Livingstone expands, places are not only where science is produced, but also where it is
encountered and disseminated (Livingstone 2003). The containment and separation of
science from society described for laboratory settings is idealised, and the production of
98
science is constantly implicated in and connected to the outside world. Field sites are
difficult to demarcate from their milieu, and boundaries between expert and lay knowledge
become equally blurred (Henke 2000; Henke and Gieryn 2008).

2.3.1.1 The Contested Spaces of Agricultural Biotechnology


The 'politics of place' in the demarcation of boundaries between laboratory and field,
between homogenous experimental and heterogeneous everyday space, between natural and
social space, have always been at the forefront of debates around biotechnology.
Biotechnologies have inscribed a series of spaces as they have moved from place to place,
defining the relationships between nature and society and between Western science and
developing world farming. While the linearity of this description is problematic, as field
trials impact product development, and commercial propagation feeds back through
processes of monitoring and surveillance, contests have accompanied each stage of this
movement.

While the development of contained spaces for research influenced efforts to clone human
insulin, wider controversy about the dangers of recombinant DNA technology emerged
when the first experiments of GMOs, notably that of ‗ice-minus‘ crops, took place outside
laboratories (Jasanoff 1995; Krimsky 2005). Placing GM crops in the field has created
novel intersections between lay and expert, everyday and experimental, public and private
space, as the places of biotechnology production also become those in which it is
encountered. Consequently, conflicts occur between assuring technoscientific quality
through situated experimentation and the agricultural assurance of food quality.

Field trials of genetically modified crops represent an attempt to enclose agricultural space
to pursue what Lezaun and Millo (2006) have described as ‗regulatory experiments‘. Such
field trials, as ‗Farm Scale Evaluations‘ (FSE) formed part of the ‗scientific‘ strand of the
UK government‘s assessment of the economic, social and scientific characteristics of
agricultural biotechnology introduced in Chapter One. They represented an attempt to
produce ‗sound science‘ about GM crops, in the form of credible, quality knowledge about
their relation with existing farming practices and crops, but also involve ―a blurring of the
boundary between laboratory and world‖ (Szerszynski 2005:182). This blurring is evident

99
in public engagement with trial spaces, and in the difficulties in establishing the scientific
sites as both sufficiently controlled and separate, and sufficiently comparable to real-world
farming. Researchers attempted to represent a range of potential effects on biodiversity,
including those of less intensive production systems than conventional agriculture.
Nevertheless, as Levidow and Carr point out, ―government advisors signalled design
weaknesses and wider issues that would still leave ‗GM crops on trial‘‖ (2007:419).

The ‗Farm Scale Evaluations‘ were set up as scientific sites, including attempts to provide a
controlled field environment using control cases and statistically representative measures
(Lezaun and Millo 2006). However, having removed the walls of the laboratory which
allow the control of experimental space and social relations, field trials are reliant on norms
of human behaviour. For trials to proceed as science, ―the farmer must strictly follow the
growing and pesticide regime, and the rest of society must keep out of the growing area‖
(Szerszynski 2005: 189). Such societal cooperation has not been forthcoming, as field trials
have been trampled and become subject to ‗ecotage‘ (Plows et al. 2004; Szerszynski 2005;
Bonneuil et al. 2008).

Field trials are the sites at which encounters between scientific and non-scientific actors and
their understandings of the places of science and agriculture. Contest arises as the meanings
attached to locations, the symbolic features of place, clash. For researchers and regulators,
field trials are firmly experimental, a scientific enclosure removed from quotidian spaces of
agriculture. For anti-GM protestors in contrast, no such enclosure exists, and GM trials
continue to exist in ‗social space‘, imbricated with the everyday practices and bodies of
food production.

Bonneuil et al. (2008) draw on the French government report by Babusiaux et al. (2003) to
suggest that field trials have moved from being conceived as a cognitive endeavour to be
preserved from lay interference, to an intrusion into social space which must be negotiated
with actors from that space. This move compromised the early integrity of field trials,
achieved through a combination of discursive and physical boundary work which
established them as socially worthwhile and biologically contained. Yet as they go on to
describe, these physical boundaries restricted the usefulness of the data derived for the

100
assessment of applications to grow the crops commercially, on a large scale, and with no
confinement measures (Bonneuil et al. 2008). As in the case of the UK FSEs, attempts to
bridge laboratory and world impact on the quality of research.

2.3.1.2 The Emerging Geographies of Pharmaceutical Biotechnology


The removal of laboratory walls in field trials and the consequent difficulty in delineating
social from experimental space threatens the quality of both the knowledge produced and of
the food system. In contrast, conventional medical biotechnology continues to exist within
the walls of the laboratory. The places of biotechnological, biomedical and pharmaceutical
production have been relatively lightly studied within STS and related disciplines.
However, two recent studies start to develop a consideration of how these places contribute
to the establishment of biomedical quality through the micro-social description of the
facilities of UK stem cell banking.

As part of her discussion of transbiology and the ‗cyborg embryo‘, Franklin (2006)
suggests that the translation of the potential of human embryonic stem cells into biomedical
goods will ‗like agriculture‘ require a new operating system to achieve a change of scale.
The facilities of the new stem cell science are central to the definition of the relationship
between these biologies, and to defining the relationship between stem cell derivation and
IVF.

The new ‗operating system‘ described by Franklin is a central part of the quality assurance
that allows the equivalence between manufactured and ‗natural‘ biologies to be established.
Quality assurance is neither an integral nor added characteristic of these biologies, but
instead involves control over their milieu. As Franklin describes, in the context of
pharmaceutical science:

―Quality is about taking away the dirt, the noise, the pollution, the pathology and the
‗junk‘ that detract from the reliability of biological function. Therefore, quality
control, while not ‗in‘ the biotic component, culture medium, embryonic cell line,
etc., is everywhere all around it – like a protective seal against contamination‖
(2006: 172)

101
Franklin‘s description echoes Knorr-Cetina‘s (1999) characterisation of the laboratories of
molecular biology, in which she suggests that the industrial processes of molecular biology
rely on the mass production of uniform products, a form of ‗biological Fordism‘ which
assures the identity of products. This in turn is based on the purification of products from
elements that threaten their uniformity, as ―keeping things uniform requires keeping things
clean‖ (Knorr-Cetina 1999: 154).

The quality of stem cell research is established through cleanliness regimes of Good
Manufacturing Practice (GMP), clean air, international standards and accreditation. These
are embedded in the design and routines of the architecture of the stem cell facility.
Franklin (2006) describes how these bespoke facilities and their predecessors are co-
produced with the ‗cyborg embryos‘ of stem cell research.

The co-production of stem cells and their places is further explored by Stephens et al. They
draw on Thrift‘s (2006) concept of ‗performative architecture‘ to capture the effect of
building design on practice within the UK Stem Cell Bank. They analyse how the
symbolism, form and practice of the spaces of stem cell banking relate ―to the pursuit of a
socially legitimate and quality-assured science‖ (Stephens et al. 2008:88).

Stephens et al. describe the role of the architecture of the stem cell bank in establishing ‗a
metaphorical image of sterility‘ through the establishment of ―physical and symbolic
boundaries‖ which separate public and privileged spaces of stem cells (2008: 88). The
symbolic and physical boundaries which establish the sterility of stem cell production are
described in rich detail which is unavoidably lost in summary. Cleanliness is established in
a step-wise movement through different rooms and through differing types of clothing.
However, it is also embodied in a series of procedures and ‗rituals‘ reflecting the pollution
beliefs of the bank. The combination of architecture and routine, of clean rooms, standard
operating procedures and traceability is used to place stem cell matter, and perhaps more
importantly, document this placing (Stephens et al. 2008).

Of particular interest for biopharming is the assertion that the UK Stem Cell Bank is
globally important in promoting a particular vision of socially legitimate stem cell science.
As I explore throughout the thesis, technological visions of technologies and of
102
biopharming are performative, and their placing will both shape and reflect this vision.
While the studies of Franklin and Stephens et al. provide a good starting point for
discussion, biopharming‘s position at the intersection between pharmaceutical and
agricultural biologies and geographies suggests the relevance of a parallel, more developed
literature which examines the role of place in understandings of quality food production.

2.3.2 Quality and Agricultural Place

Lay publics and experts draw on a range of material characteristics in attributing meanings
to food. In addition, they draw on spatial proxies (Eden et al. 2008) related to the places of
food consumption and production. Murdoch (2005) distinguishes between topographical
and topological networks of food. Descriptions of topologies have emerged within both
STS and agro-food geography. Topology is the mathematical study of relations, but as used
by authors such as Law and Mol (2001) has been used to describe ‗network space‘. For
Law and Mol (2001) the distinction between topology and topography is used to separate
networks from ‗topographical‘ or Euclidean space. In this thesis, I follow Murdoch (2005)
and the work of Eden et al. (2008) introduced above, in using topologies to describe
particular networks of food production which constitute relations between the locations of
consumption and production.

The topologies described by Murdoch are those such as the organic cheese production
discussed by Murdoch et al. (2000), in which quality is constructed through the embedding
of production in relations between producers, consumers, animals and locales. Within these
‗topological spaces‘ of food, consumers are encourage to acquire knowledge about the
provenance of food and engage with foods in the sensory ways described by Roe (2006).
As such, the black boxes described by Eden et al. (2008) above are opened as potentially
distant consumers (in a Euclidean sense) are drawn into ‗discrete cultural contexts‘ of local
production. At the risk of homogenising a diverse range of alternative food production
networks, the value of food within them, for both producers and consumers, and its
perceived quality are linked to embedding in particular relations of production.

103
Alternative food networks relate the qualities of foods to health, environmental, nutritional
and ethical characteristics, often through association with particular production locations.
Sonnino and Marsden (2006) suggest that these qualities are defined in opposition to the
perceived characteristics of conventional agriculture. In these terms, they describe quality
in food as a ‗multidimensional concept‘ involving such features as an identifiable place of
origin, traceability, nutritional value and the aesthetic attributes described in the previous
section. As such, quality acquires meaning only with within specific production–
consumption relationships (Sonnino and Marsden 2006).

Murdoch (2005) suggests that these topological networks of production are typified by the
‗Slow Food‘ movement, which emphasises placed foods, including through certification
such as that provided by the Appellation d‘origine contrôlée (AOC) system for wines.
Indicators of quality such as the AOC designation are tied to particular terroirs or locales of
production, and to specific local technical systems and savoir faire/know-how (Murdoch
2005). These indicators allow consumers to approach the origins of their food, creating new
relationships between them and producers.

Murdoch contrasts these topological, relational designators of quality with the


topographical networks of ‗fast food‘, particularly McDonald‘s restaurants (see also Star
1991). Similarly, Sonnino and Marsden suggest that:

―[T]he contemporary food sector is increasingly bifurcating into two main ‗zones‘ of
production: standardized, specialized production processes responding to economic
standards of efficiency and competitiveness on the one hand; localized, specialized
production processes attempting to trade on the basis of environmental, nutritional,
or health qualities on the other‖ (2006:183)

Within the networks of McDonald‘s production, the quality designations that tie production
to terroirs are replaced by universal standards and the coordination of heterogeneous
elements to homogenise and ‗black-box‘ food production. Thus the process for cooking a
burger or an apple pie becomes ‗immutably mobile‘ (Latour 1988) and can be maintained
across McDonald‘s branches, providing standardised products using specialised, computer
controlled machines. This standardisation and homogenisation of food production ensures a
consistent quality of product and enables ‗topographical‘ networks of production. As
104
Murdoch expands, these topographical networks involve new relations with consumers, in
which attention is diverted away from the qualities of foods themselves, and towards
characteristics such as price or speed. Layers of ‗symbolic artifice‘ (Murdoch 2005: 175)
distract attention from the foods themselves.

Enacting the topographies of McDonald‘s involves standardising the behaviour of human


actors within the space of the company‘s restaurants. In order for standardised products to
be produced, it is necessary for the correct protocols to be followed, and for the behaviour
of workers within the restaurant space to be closely regulated. It is the combination of
physical and procedural control that allows the establishment of topographies of fast food.

Murdoch (2005) extends his discussion into the spaces of agriculture, suggesting the
standardisation and homogenisation of the restaurant is accompanied by that of agriculture.
Hence the ability of McDonald‘s to produce a standardised product involves not only
control and disciplined spaces of consumption and food preparation, but a disciplining of
agricultural nature through attempts to produce a consistently sized and shaped potato.

2.3.3 Combining the Spaces of Food and Pharmaceuticals

The protocols and standardised spaces of conventional, topographical food production


described by Murdoch (2005) echo the biomedical quality regimes described by Stephens
and Franklin above, and suggest that the movement between some places of food and those
of medicine may not be so great. As does the stem cell bank, McDonald‘s restaurants
excise extraneous matter from production to establish quality through the removal of the
dirt, noise and pollution.

Bonneuil et al. (2008) suggest that the extension of conventional agriculture through GM
may bring these networks even closer. They draw comparisons between the spaces of
agricultural biotechnology they describe and those of pharmaceuticals. However, they
suggest a different form of pharmaceutical space to that described by Stephens and
Franklin. For Bonneuil et al., the conception of field trials as regulatory experiments, with
experimental norms, statistics and market approval procedure constitutes them as an
agricultural equivalent to clinical drug trials.
105
Similar parallels are present in Levidow and Carr‘s (2007) discussion of regulatory
approaches to GM which involve post-marketing monitoring. In line with the discussion
above about the difficulties of delineating experimental space, they suggest that this
constitutes ‗society as experiment‘ (cf. Krohn and Weyer 1994). Such monitoring brings
the post-consumption assessment of food together with that of pharmaceuticals, whose final
stage of trialling involves such monitoring. In this sense, society is already an experiment
with the release of every novel pharmaceutical.

However, the convergence of pharmaceutical and agricultural space may be rendered more
complicated by the valorisation of place in food production described in section 2.3.1. For
both producers and consumers, recent years have seen a ‗turn to quality‘ in food and the
emergence of ‗alternative food systems‘ (Sonnino and Marsden 2006; Murdoch 2005). The
existence of powerful alternative food networks raises potential problems for the
development of biopharming, as it has for first generation genetically modified crops.
These alternative food networks involve a differing conception of and role for place to the
‗topographical‘ networks of McDonalds and stem cells. Instead, they are topological,
relying on the existence of relationships between producers and consumers. The ways in
which quality of food is asserted within these topologies appears to differ markedly from
that of biomedical quality asserted through GMP and clean air. I explore the consequences
of this for biopharming in more detail in Chapter Six.

2.4 Summary and Conclusions

In this chapter, I considered the introduction of biopharming as a question of ‗placing‘, of


the establishment of temporal, ontological and spatial relations between novel, plant made,
pharmaceuticals and the foods and medicines with which we live. In the empirical chapters
of the thesis I develop these arguments through the focus group discussions and interview
transcripts which form the data for the thesis. Each chapter picks up and contributes one of
the ‗placings‘ described here.

In the first section, I considered how narratives of biopharming may be used to constitute it
as a technology of pharmaceutical potential and shape the form through which that this
106
potential is actualised. I argued that technologies are placed through hope and affect, rather
than effects. I suggested that these are not only a question of narrative, but of imagined
geographies. I pursue this discussion in Chapter Four, in which I examine how the future of
biopharming is narrated by focus group participants and biopharming experts. I examine
how biopharming is placed within narratives of pharmaceutical and agricultural research
and development.

In section two, I addressed how the novel ontologies created by biotechnologies are placed
through association with existing products. Combining work on food consumption with
discussion of regulatory approaches to substantial equivalence, I suggest that for both lay
people and experts, the relational materialities of food contribute to judgements of safety
and quality. I suggested that there is a lack of similar research in the case of
pharmaceuticals. In Chapter Five, I extend this discussion to compare and contrast
biopharming‘s encounters with the matters of pharmaceuticals and foods.

Finally, in the third section of this review, I have brought together two bodies of work to
examine the role of place in the development of biotechnology. The first addresses the role
of place and space in the production of scientific knowledge and of pharmaceuticals, while
the second considers how similar questions of locality arising in agricultural production
play a role in determining food quality within ‗topological‘ networks. I suggest that the
places of technoscience are not only backdrops for the production of knowledge, but are
constitutive of the quality of the scientific product, and as importantly understandings of
this quality and expectations of its worth. I develop this empirically in Chapter Six, in
which I investigate the role of place in pharmaceutical production processes, and consider
equating the pharmaceutical facilities and architectures described by Stephens et al. (2008)
completes the ‗placing‘ of biopharming‘s promise.

In the subsequent chapter, I consider how the theoretical and empirical concerns I have
introduced here can be addressed methodologically through an approach that combines
expert and lay narratives, ontologies and places of biopharming.

107
CHAPTER 3: QUALITATIVE METHODS AND
PLACING BIOPHARMING

The introduction of novel biotechnologies involves not only the achievement of technical
success, but also public and financial support. This requires the promise of new
technologies to be understood and ‗placed‘ in relation to existing ones. The methodology
adopted in this research attempts to capture the heterogeneity of this process, from the
discourse of publics and researchers through a series of interviews and focus groups, to the
texts of research and regulatory documents, to the products of foods and pharmaceuticals
and the materialities of plant and human biologies.

The purpose of the thesis is not to present ‗public attitudes to biopharming‘ in isolation, but
to bring together and compare public and expert perspectives on the placing of biopharming
within a social research setting. While much effort has gone into study of public
perceptions of new biotechnologies (for example Gaskell 2004; Gaskell 2007; Marris et al.
2001), the voice of experts still tends to be dominant in situations where the introduction of
these technologies is described. Despite her emphasis on public knowledges and ‗civic
epistemologies‘ Jasanoff (2005) focuses almost entirely on expert and ‗elite‘ actors in
discussing how novel biotechnologies ‗make a place‘ among existing societal narratives in
the UK, US and Germany. Yet the narratives, artefacts and places among which new
biotechnologies emerge are part of everyday life as much as they are of the world of
biotechnology policy and science. In this chapter I consider how the multiple discursive and
material repertoires through which publics and experts make sense of novel biotechnologies
can be examined.

The combination of experts and publics here echoes the work of researchers such as Kerr et
al. (1998) on medical biotechnologies or Marris et al. (2001) on agricultural
biotechnologies. Rather than staging a direct dialogue, this study follows prior research in
situating researchers and lay perspectives side by side within their social context. The
studies of both Marris and Kerr consider public perceptions of novel technologies, but
concentrate on expert perceptions of the public. In contrast, the research presented here
examines expert and public placings of the same technology, with the aim of describing
convergence and divergence between them. Moreover, it builds on Michael‘s discussion of
a ‗heterogeneous PUS‘ in attending to the technologies, materialities and places through
which encounters between experts and publics occur.

The selection of qualitative or quantitative methodologies reflects particular intellectual


traditions and embodies particular models of the nature of the person, of the role of
knowledge, the structure of society and the purpose of politics (Irwin and Michael 2003)10.
Qualitative approaches allow the researcher to examine the narrative structures through
which people make sense of the world and examine their engagements with the materials
and spaces of everyday life. As they avoid, to some extent, the imposition of the analyst‘s
categories on data, they are especially useful in the exploration of science and technology
in people‘s own terms, whether lay or expert. They are especially appropriate for
examining the lay epistemologies described in Chapter One and to explore how scientific
questions are situated within their social and cultural context (Irwin and Michael 2003).
This approach emphasises that lay publics not only possess knowledge, but to know how
they know, that they assess the institutional contexts and trustworthiness of different
knowledge claims, and that they relate these to the knowledges and artefacts of everyday
life.

Davies and Dwyer, in a series of reviews of the state of qualitative methods in human
geography (2007, 2008) suggest a change has occurred in how qualitative social science
research is conceived and carried out, and that this has been accompanied by change in how
such research is used to make claims about the world. They suggest there has been a
rejection of singularity in social science research methods. Rather than the use of empirical
research with the assumption that it will generate clarity, precision and reduce uncertainty

10
Although qualitative methods are conventionally associated with ‗contextual‘ approaches to the public
understanding of science, they can be applied in a ‗deficit‘ manner, and quantitative methods can be used to
examine lay knowledges (Sturgis and Allum 2004).
109
and ambiguity in our understanding of the world, instead it has to be recognised that all
research is performative and helps to enact particular realities.

For an emerging and non-stabilised technology such as biopharming, the performative role
of social science research is important. Davies and Dwyer (2007) suggest that researchers
must attend to what it means to engage with the world through research – methodologically
and through our interpretations and ethical aspirations.

In these terms, the ethics of undertaking research on a promising medical technology are
particularly important. Biopharming promises transformations in the production of
pharmaceuticals and in their availability to the developing world. By offering a critical
perspective on biopharming, my work may either add to the hype or generate
disenchantment by creating its own performative futures and expectations. However, as the
purpose of the research is to present a discussion of biopharming in its social context, this is
unavoidable and by no means negative. The identification of intrinsic and extrinsic barriers
to the contribution of biotechnology to the delivery of equitable healthcare is both a useful
and ethical intervention into the biopharming debate.

Within the current research, I have attempted to approach this engagement with broader
biopharming research by publishing on ‗public attitudes to molecular farming‘ at an early
stage (Milne 2009). By feeding the results of my research with publics into biopharming
research, I hope to contribute to the development of the technology as ‗socially robust‘. In
particular I intend that the thesis will contribute to moving understandings of public
attitudes to biotechnology beyond ‗pro-‗ or ‗anti-‗ positions.

By providing alternative representations of public concerns that resist polarisation, social


science can inform a more broad-ranging ‗upstream‘ debate about the social purposes that
science should serve. (Kotchetkova, Evans, and Langer 2008). By placing expert and lay
perspectives alongside each other, I attempt to contribute to efforts to incorporate public
and societal framings at an earlier stage of technological development.

A range of qualitative methods were used in the study: six repeated focus groups were held
in London and Northern England, to survey the discursive landscapes of medicine, food

110
and biotechnology, and to place molecular farming within this. The approach adopted
involves tracing the relationship between medicines and foods from their quotidian and
mundane forms, through recent intersections with biotechnology to the prospective form of
plant made pharmaceuticals. Throughout this, public voices are joined by a series of
researcher and regulatory interventions and framings. These were examined through semi-
structured interviews with researchers and a review of research papers. Together, these
methods contribute to understanding the ‗placing‘ of molecular farming in the UK.

3.1 Focussing on Groups

The first strand of the research approach endeavours to capture the placing of biopharming
by the ‗lay public‘, those who have everyday knowledge of food, medicine and
biotechnology, as opposed to the expert or scientific knowledges approached through
interviews and review papers.

To do this, six focus groups involving a total of 35 participants were constituted from pre-
existing groups in London and Lancashire. Each group met twice, and was provided with
information about biopharming between meetings. I discuss the specific logistical and
procedural choices involved in this research in more detail below. However, I first consider
the reasons for using focus groups to study public placings of biopharming, and describe
some of the diverse ways in which focus group methods have been adopted.

3.1.1 Introducing Group Methodologies

Morgan defines groups as a ―research technique that collects data through group interaction
on a topic determined by the researcher‖ (Morgan 1996:6). As recently as 1997 they were
described as under-used in social science research (Gibbs 1997). However, their use has
become well-established in academic social science research over the last 20 years, such
that Lezaun can now comment that ―provoking a conversation among a small group of
people gathered in a room has become a widespread way of generating useful knowledge‖
(Lezaun 2007:130).

111
The roots of focus group research date to the 1950s and investigations of the effects of war-
time propaganda. However there is considerable variation in approaches to the conduct of
focus group research, and differing conceptions as to what distinguishes typologies of
group discussions. The early history of focus groups research involved a movement from
academia to market research. Over the last 25 years, focus groups have re-entered the
academy, albeit often with a market research approach to methodology and analysis still
attached (Cunningham-Burley, Kerr, and Pavis 1999). Their conduct ranges from highly
moderated, constrained, one-off discussions approximating market research groups through
to ranging discussions with low levels of moderation over a series of meetings, such as
Burgess, Harrison and Limb's (1988a, 1988b) research drawing on the different tradition of
therapy groups.

Groups allow participants to generate their own questions, frames and concepts to a greater
extent than methods such as surveys, and pursue their own priorities according to their own
terms of reference (Kitzinger and Barbour 1999). As such, small group work is excellent
for exploring people's knowledge and experience in depth and gaining understandings of
how and why people think about subjects. They are carried out using participants' own
vocabulary and own forms of communication, including informal, day-to-day interaction
such as argumentation or the use of anecdotes. As such they are useful when it comes to
investigating what participants think ―but they excel at uncovering why participants think
as they do‖ (Morgan 1996:25).

Focus groups are a more ‗naturalistic‘ form of research method than the questionnaire
survey, while allowing the researcher to direct discussion in a manner impossible in
conducting participant observation or more ethnographic work. As they explore how people
themselves categorise and make sense of their own experience, they can allow the
researcher to avoid imposing their own understandings (Marris et al. 2001). In work on
environmental risk, it has been suggested that groups are thus useful for obtaining a
‗relational‘ perspective, exploring both how people form attitudes through interaction,
rather than these being something held pre-formed within, and ―the connections between
how people experience, define, think, feel and represent risks in relation to wider debates‖

112
(Waterton and Wynne 1999:133). In the context of ‗placing‘ biopharming, this use of focus
groups to situate discussion among the everyday is particularly pertinent.

The group process allows the researcher to observe many of the processes that occur as
people actively form attitudes towards subjects not often addressed (Cunningham-Burley,
Kerr and Pavis 1999). This raises questions about the role of the facilitator and the means
through which these subjects are introduced. Nevertheless, in comparison with individual
interviews, groups provide a forum for the sharing and testing out of views which illustrates
the dynamics of thought, speech, and understanding between subjects (Cook and Crang
1995; Burgess et al. 1988a). Focus groups are thus particularly well suited to explore public
attitudes in contexts where participants have little prior exposure to a topic. As Sturgis and
Allum (2004) describe, other methods such as surveys encounter significant problems in
dealing with technologies with which participants have little familiarity. To circumvent
these problems, they suggest that studies of low-salience topics, which biopharming
undoubtedly is, should include in depth qualitative aspects.

As evidenced in the citations throughout this introduction, while focus groups continue to
be used extensively in social science research, not least in the public understanding of
science, much of the practical guidance in their conduct is now at least a decade old.
Hopkins (2007) also notes that even in 1999, Kitzinger and Barbour were concerned that
social scientists were ―in danger of uncritically adopting market researchers‖ models of
such research rather than adapting and expanding them, taking into account our own
purposes and theoretical traditions‖ (2007: 529). While there is undoubtedly a risk that
basic focus group methodologies are adopted uncritically and formulaically, the evidence of
recent work in both research and deliberation around the public understanding of science
suggests a diversity and originality of applications (Burgess et al. 2007; Armstrong et al.
2006; Einsiedel and Medlock 2005; Michael and Brown 2005a). Indeed, it may be the case
that the ‗modified focus group methodology‘ is now as common as standard ‗one-off
meetings‘.

Hopkins (2007) suggests that the lack of significant recent consideration of focus group
methodologies is not evidence of an absence of development, but rather that researchers

113
choose not to reflect at length on methodological questions in their writing. Nevertheless,
the diversity of group methods begs the question of whether the advice provided for the
practice of one-off groups is relevant to modified methodologies. For example, Burgess,
Harrison and Limb‘s (1988a, 1988b) work with ‗in-depth‘ groups draws on a tradition of
group therapy, and involves particular considerations in the forming and winding down of
group discussions that are less relevant to one-off or ‗short-repeat‘ groups.

One problem with group methodology, particularly in contexts where participants are
discussing novel or unusual questions, is that there is a shared assumption that the purpose
of the discussion is to display opinions to the facilitator of the group (Myers 2000).
Opinions consciously put on display are not inherently of less validity than if they had been
kept to themselves. However this does raise questions about the extent to which focus
groups can be said to produce 'naturally occurring' data and highlights concerns about the
identity of research subjects in qualitative group methods. Focus group methods, along with
other social science techniques act to constitute communities and subjectify participants.
This attunes us to the limitations of focus group methods, and recalls that

―we, with our social scientific procedures, are a part of the production of
understanding and of the public... These techniques, procedures, categories, and
discourses serve in the making of people: they comprise some of the resources
through which lay people come to perform themselves in relation to one another,
science, and social science‖ (Michael 2002:374).

Laboratory places of science attempt to delineate and control the natural within a confined
setting, but must sufficiently approximate particular natures. In a similar fashion, the focus
group creates a delineated but recognisable experimental setting for research into 'society'
(Lezaun 2007), neither equating the contained controlled space of the laboratory or
interview room, nor that of the field or ethnographic site. Instead, in contrast to the defined
limits of surveys, the focus group, it can be argued, represents the social science equivalent
to the ‗field trial‘, in which society itself is experimentalised (cf. Krohn and Weyer 1994).

Focus group participants in this vein become experimental subjects. Hence the description
above of groups as ‗naturalistic‘ rather than ‗natural‘, as the interaction they capture is
inevitably artificial, but derives its value from its approximation to un-moderated, everyday
114
discussion. As I describe in the following section, these considerations provide an
important starting point in selecting who and how to recruit for the research.

3.1.2 Who to Recruit? Naturalism and Pragmatism

Having established the value of a focus group approach for the exploration of publics‘
relations to new technologies, the first concern in the development of the current research
was with the recruitment and constitution of the groups. In the market research tradition,
attempts are made to simulate an experimental setting by including participants who have
never previously met. In this vein, some moderators emphasise that the value of group
discussions is their ability to extract information in ‗a radical experimental intervention‘
(Lezaun 2007). In contrast, others consider focus groups as a less overtly experimental
approach in which the value of data emerges from considering how participants might
discuss the focus of group discussion in everyday life (Morgan 1996; Kitzinger 1995).

Large scale studies such as Marris et al. (2001) and Armstrong et al. (2006) often use
agencies to recruit participants. However, given the financial restraints of a PhD, such
methods were not available in this study. Fortunately, there is significant evidence in
support of the use of ‗pre-existing groups‘, or groups of individuals who already know each
other, as an alternative to those constituted for the first time by the researcher. This
approach parallels that of previous research in the ‗public understanding of biotechnology‘,
particularly that of Michael and Brown on risk identities in the context of
xenotransplantation (2005a), in which they used a series of 11 groups constituted from
sports clubs, colleges and community organisations. Similarly, Coyle and colleagues
(Coyle and Fairweather 2005) used school PTAs to contact a range of participants in their
study of public attitudes to biotechnology in New Zealand. Green et al. (2003) also used
groups of individuals who already knew each other.

Kitzinger (1995) argues that research using groups formed from pre-existing groups may
generate fragments of interaction which approximate to 'naturally occurring data' through
tapping into what Khan and Manderson (1992) refer to as 'natural clusterings' representing
an approximation of the sources of material, information and advice which a member of the

115
group might draw on. Kitzinger (1995) suggests that the fact that people know each other
has the additional advantage that they are able to relate each others' comments to actual
incidents in their shared lives. In their work on public understandings of genetics, Kerr and
colleagues also draw on pre-formed groups, suggesting that they ―are more easily
accessible and more relaxed about discussing sensitive and complicated topics than groups
of strangers‖ (Kerr, Cunningham-Burley and Amos, 1998:44)

Holbrook and Jackson (1996) suggest that forming groups from participants who know
each other may reduce the anxiety participants might feel in joining a group of strangers.
As such, it may encourage people to participate who may have declined had other research
methods are used, and decrease the difficulty in joining in the conversation. Connected with
this latter point, Holbrook and Jackson suggest that ―at times it felt as though we were
'eavesdropping' on conversations that could have occurred without our presence‖ (1996:
139). Indeed, in the groups convened for this research, discussion of foods and medicines in
particular often took this form of free-running discussion that could have occurred without
my intervention. In contrast, when groups were presented with biopharming, as described
below, discussion became much more stilted. Nevertheless, in attempting to examine how
biopharming is ‗placed‘ among more everyday technologies, this approximation to
‗everyday‘ interaction is potentially invaluable.

However, in the research it also became clear at times that the use of pre-existing groups,
particularly those who meet regularly, is not without its problems. The research process can
both display and reinforce existing hierarchies and power relationships and imbalances
within the group (Kitzinger and Barbour 1999), such that some participants attempt to
dominate group discussion. Unlike in groups convened solely for the purpose of research,
the ability of the moderator to intervene is diminished when dealing with relationships and
group dynamics that pre-exist the research context.

The role of the moderator is thus both potentially easier and more complicated than in focus
group research with one-off, newly formed groups. While it is easier to obtain discussions
that are free-flowing and naturalistic, facilitation needs to remain aware of the groups
dynamics. In the current research, this represented a particular problem in two groups

116
(notably C and E, introduced below) in which some participants were noticeably dominant.
As such, a more challenging approach to facilitation than that adopted in other groups was
required to enable other group members to participate.

Moreover, not all existing groups are equivalent: the degree to which a group might at any
stage approximate 'naturally occurring' discussion is reliant on the type of group, the
frequency with which they meet, and their reasons for meeting. For example, in their study
of tropical diseases Khan and Manderson (1992) drew on groups consisting of family and
close friends in small communities. In the present study a variety of groups was used, as
shown in Table 3.1 in the following section. These ranged from those well-acquainted with
each other (particularly groups C, D and F), to those less well acquainted (particularly
group A). In addition, the groups that met were often only a subgroup of a larger
community. As such, a participant might know some of other group members better than
others. In practice however there was little difference in the interactions between group
members, although those groups best acquainted were often able to challenge each other on
the difference between their reported and actual behaviour. This suggests significant
advantages for the use of existing groups, as described by Holbrook and Jackson (1996).

3.1.3 Recruiting the Research Groups: Heterogeneity, Homogeneity and


Hard Work

Having established that pre-existing groups represented a potentially productive line of


recruitment, a range of existing groups was contacted to include a variety of participants.
Some, such as Einsiedel and Medlock (2005) try to ensure that their groups are in some
way ‗representative‘ of the wider population, although it is difficult to achieve any genuine
representativeness in such a small sample (Morgan 1996). As Kitzinger and Barbour point
out, focus group samples do not hope to achieve a statistically representative sample, but to
―encompass diversity and compose a structured rather than random sample‖ (Kitzinger and
Barbour 1999:7). As such, recruitment aims to ensure that there are no significant
exclusions from research. For example, the PABE study (Marris et al. 2001) attempted to
ensure that a comprehensive range of views were expressed by including an explicitly

117
‗socially excluded‘ group, in their case a group who were unemployed with low income
and education.

The groups convened for this research included a diverse spectrum of participants, and I do
not feel that there were any significant exclusions from the research. The purpose of focus
group research is not to capture a picture of all possible opinions on a topic, but to provide
a detailed image of some of the most pertinent. To this end, I decided to recruit six groups.
As Morgan (1996) advises, more than five groups rarely provide novel insights, as a
measure of ‗saturation‘ is achieved. However, holding six groups allowed an equal division
between urban and rural settings.

While some, such as Beckwith et al. (2003) have worked with specifically interested
groups, the intent in my research, as in the PABE study (Marris et al. 2001), was to involve
a range of groups from among the UK public who do not have a specific vested interest in
pharmaceuticals, biotechnology or agriculture. While Group E was formed from the
members of a gardening club, they were not necessarily involved in the production of food
or commodity crops.

As Morgan (1996) describes, the ideal of focus group recruitment is to ―create a variety of
internally homogeneous groups that capture a wide range of potentially distinct
perspectives‖ (1996: 37). While maximum heterogeneity is thus sought between groups to
provide a range of responses, homogeneity is valued within groups. As one guide to focus
group recruitment points out "most handbooks advocate a relatively homogeneous social
group" (Cook and Crang 1995:57). Homogeneity within groups increases the richness and
depth of group discussions as people become more willing to share experiences and
understanding with those like them. This homogeneity was achieved in the current research
through the use of pre-existing groups, who had at least membership of the group in
common, along with shared interests, work or concerns.

The focus group meetings for the research took place in London and Lancashire,
representing urban and rural regions. This divide provided increased heterogeneity between
groups, allowing a broader spectrum of views to be heard. In addition, it was thought that a
rural location might influence group discussion, particularly about food. Potential groups
118
were identified using directories in libraries, community centres and online, both through
indices such as Lancashire Lantern, run by Lancashire county council, and through internet
searches using Google. It is generally acknowledged that recruitment of focus group
participants can be time consuming, especially if the topic under consideration has no
immediate benefits or attractions to participants (Gibbs 1997). To encourage people to
participate, incentives such as expenses, gift vouchers or presents are often offered. As the
groups in my research were to meet on two occasions a week apart, requiring a significant
time commitment, these difficulties were further exacerbated.

My first recruitment tactic was to send potential groups an introductory letter and a flyer
explaining the research. This was followed by a phone call a week later11. Participants were
initially offered a £20 inducement to attend two focus group meetings to discuss ‗the future
of food and medicine‘. As can be seen in Appendix I, while the flyer mentioned that the
research would be considering biotechnology and genetics, it did not specifically mention
biopharming. This method of contacting groups was spectacularly unsuccessful. In fact,
only one of the six groups was recruited in this way (Group E) despite contacting 50
societies, organisations and clubs by post or email. One reason for the lack of interest may
have been the commitment required from participants: an hour and a half on two occasions.
Similar recruitment problems are described by Holbrook and Jackson (1996) who also had
little response to letters or posters in their attempts to recruit groups for three meetings to
discuss experiences of shopping.

After several months of unsuccessful attempts to recruit, I raised the incentive to £30, and
reconsidered my strategy. While I carried out a pilot group with undergraduate students, I
was keen to extend both the scope and relevance of the research by contacting a spectrum
of people. In their elaboration of their recruitment travails, Holbrook and Jackson (1996)
emphasise the importance of contacting potential participants in person or through a trusted
gatekeeper, or ‗snowballing‘ (Burgess 1996). I therefore attended meetings of groups to
recruit people in the places where they meet to routinely talk and socialise.

11
The flyer and letter are reproduced in Appendix 1

119
Following this change of strategy, I had significantly more success. Five groups were
recruited through personal contacts or through others who knew of, or were part of the
groups themselves. These individuals acted as gatekeepers and as recruitment agents. For
example the widows‘ support group in Lancashire (D) was contacted through the friends of
an elderly relative, while the Groups B and F were also approached through existing
acquaintances. Other groups were recruited in person during their regular meetings. The
church group was contacted at its weekly coffee morning, while the group formed of
cricket club members was contacted at their weekly practice session. The problems
encountered in recruitment re-emphasise the importance of personal contact in initial focus
group recruitment, whether by the researcher or a trusted gatekeeper. However, once the
groups were recruited, not a single participant dropped out between the first and second
group meetings.

The eclectic forms of recruitment resulted in the formation of six heterogeneous but, to a
large extent, internally homogenous groups from a range of existing relationships.
Ultimately, the diversity of participants that resulted from a somewhat tortuous recruitment
process enriched both the final research and the research process itself. Advice on the
number of participants per group varies, from between four and eight (Bedford and Burgess
2001) to between eight and twelve in market research literature. Among the contributors to
the edited collection by Kitzinger and Barbour (1999), group membership ranges from
three to six. In my research, the size of group varied between four and eight, as presented in
figure 3.1. I found that five or six participants was the ideal number to encourage lively
discussion.

120
Figure 3.1: Focus group constitution

Group
Group Age Gender Location Type of Group
size

A Young (<40) Male Urban Cricket club 7

B Young Female Urban Work colleagues 4

Methodist Church
C Older (>40) Mixed Urban 5
group

Widows‘ support
D Older Female Rural 6
group

E Older Mixed Rural Gardening club 8

Teachers (but not


F Young Mixed Rural 5
colleagues)

Focus groups can be held in a range of locations (Gibbs 1997), and it is important to
consider how these locations affect group discussion (Hopkins 2007). As with recruitment,
I adopted an approach which combined theoretical and pragmatic considerations. In using
pre-formed groups, the question of where to hold meetings is often taken out of the
researcher‘s hands. For example, the gardening group (E) and Methodist group (C) have
established meeting places, and it was possible to co-opt these locations for the purpose of
the research. It was not possible to hold group meetings at the same time as the groups‘
normal meetings – for the Methodists their twice weekly coffee morning, at which I met
and recruited them, is also a drop-in centre, and as such has other roles to play. For group
A, the cricket club, I hired a room in the school in whose sports hall indoor practice took
place, and participants were thus able to come to the group session without leaving the

121
premises. Group B met in a room at their workplace, while Groups D and F met at the
houses of a group member.

Each focus group location had its advantages. Both groups held at participants‘ homes
featured particularly lively discussions, although whether because participants felt more
comfortable or were particularly lively groups is impossible to say. In addition, they
required less travelling, and guaranteed attendance, for at least some participants. However,
in Group D a break in the discussion was extended by the host leaving to make tea, so time
was lost for group discussion. Such problems are not encountered in using more ‗neutral‘
territory in which it is easier for the facilitator to direct the schedule of group discussion.

3.1.4 Practice of Groups

Focus groups with pre-existing groups represent a compromise in qualitative research


methodology. They sit between observational techniques such as ethnography, in which the
researcher attempts to observe everyday practice and discourse in its milieu, but which are
of little use in considering topics beyond these everyday activities, and the more
interventionist and experimental settings of the market research ‗group interview‘ and
individual interviews.

Holding two group meetings allowed discussion of biopharming to take place in the context
of participants‘ existing views about food and medicine, and for information about the
novel technology to be provided. Each meeting lasted for an hour and a half. The protocols
used to prompt and guide group discussion are reproduced in Appendix III. These were
trialled and refined in a pilot study using a group of seven undergraduate students in the
STS department at University College London.

The first meeting focussed on general attitudes to food, medicine and to biotechnology.
This session was designed to explore the ways in which people relate to the production and
provision of food and medicine, and how decisions about these are made in their day-to-day
lives. This discussion provided a basis for further discussion when molecular farming, a
topic with which the groups were unfamiliar, was introduced. This first group featured a
relatively free, wide ranging exploratory discussion of participants understandings of food
122
and medicine, with the direction of discussion left much to the determination of the group
itself.

As Michael (2002) points out in describing the ‗heterogeneous public understanding of


science‘, focus groups are not only a methodological tool but a semiotic-material
assemblage of spaces, objects, people and performance. In many ways, the material aspects
of the group discussion are as important as the participants themselves, as science is
‗prehended‘ by participants ―not only through representation but also in everyday material
encounters with…technology and nature‖ (Michael 2002: 373). It was thus important to
capture how the everyday materialities of foods and medicines are ‗prehended‘ within
group discussion.

Here the research of Roe (2006) is again of relevance. As she describes, by presenting
participants with ‗ordinary' carrots, knowledges are expressed which touch upon the non-
discursive and non-reflexive level of human experience. Prompting discussion with
conventional foodstuffs thus directs participants discussions through the everyday
aesthetics and practices of eating, rather than starting with the abstract considerations of
attitudes and opinions that characterises studies such as GM Nation? (DTI 2003).

Moreover, Green et al. (2003) suggest that there is a divergence in discussion of food
between ‗rational‘ decisions made by consumers about ‗kinds of foodstuffs‘ and the
everyday decisions about particular items. As such, a range of foods was provided as a
prompt for discussion, to capture this distinction between general and particular
discussions, and the material interactions with everyday natures and technologies. Each
group discussed a Ginster's Cornish pasty, a packet of probiotic yoghurts, an apple (either
Granny Smith or Braeburn), and a packet of tricolore pasta.

The props were selected to capture a range of concerns about food based on the theoretical
perspectives introduced in Chapter Two. For example, as can be seen in Figure 3.2, the
Cornish pasty is replete with imagery intimating its quality and trustworthiness in the form
of an image of the pasty contents, the motto ‗Cornish through and through‘ and a range of
geographical and traditional symbols. The pasty is linked to Cornwall through the
background image and the Cornish flag on the logo. Cursive ‗olde-world‘ handwriting on
123
the label and product description completes the image by conjuring images of traditional
signs.

The Cornish pasty, with its explicit evocation of local production, was used to explore the
importance of ‗place‘ in food production, while the examples of probiotic yoghurts and
apples opened up a space for discussion of healthy eating.

Figure 3.2 The Ginster’s Cornish Pasty

The extent to which such props begin to capture everyday interaction with foodstuffs can be
illustrated by their relationship with other foods present in group meetings. Groups were
also provided with a range of snacks for their own consumption, often placed on a table in
the centre of discussion. In a number of groups, the distinction I had envisaged between
‗props‘ and ‗snacks‘ collapsed, with the latter drawn into discussions around healthy eating
or food production, and the former eaten during the meeting.

The second stage of discussion extended this approach by exploring similar questions for a
range of over the counter and herbal medicines. These included arnica, a herbal treatment;
ibuprofen, a synthetic pharmaceutical; sticking plasters, to broaden discussion beyond
internal medicines and aspirin, a 'modern' pharmaceutical initially derived from South
American willow, to encourage participants to explore the provenance of medicines. This
discussion of material pharmaceutical props allows the ‗ideal type‘ medicine described by

124
Marris et al. (2001) to be grounded in the same types of quotidian knowledge described
above. Again, the range of medicines was chosen to provoke or evoke particular types of
argument, in particular to consider the relationship between herbal and synthetic or
industrial pharmaceuticals, in advance of the discussion of biopharming the following
week. The introduction of pharmaceutical props after foods was intended to explore the
relationship between the terms in which foods are discussed, on which a significant
literature exists, particularly in terms of place and quality, and those in which
pharmaceuticals are considered. One immediate difference emerged from groups‘
approaches to the products. While food props were eaten as readily as the snacks provided,
the pharmaceuticals remained untouched.

Having initiated discussion through concrete, tangible and familiar items, I then moved the
discussion to consider biotechnology. Participants were asked what, if anything, they knew
and felt about a list of applications, including GM crops and food, xenotransplantation,
pharmacogenetics, genetic testing, gene therapy and stem cell research. This discussion
generally covered the last twenty minutes to half hour of the session, and allowed groups to
relate these applications back to their earlier discussions. Most groups spent around half of
this time discussing GM crops. No props were used in this section of the discussion.
Instead, I asked participants to relate their knowledges, perceptions and concerns about
biotechnology back to the earlier discussion of food and medicine. Consequently, many of
the same discussions re-emerged, refracted through the novel technologies. Throughout the
group process, I emphasised that participants were not being ‗tested‘ on their knowledge of
biotechnology or of molecular farming.

An obvious problem with the use of focus groups to generate ‗naturalistic‘ data is that
subjects such as biopharming, which are of low salience, are unlikely to feature in everyday
discussion, and definitely not in the form of a set discussion such as that undertaken here.
The amount of information to provide to participants about biopharming was thus an
important consideration in the development of the group methodology. As discussed by
Einsiedel and Medlock (2005), molecular farming presents difficulties for focus group
analysis due to its novelty. Kerr et al. (1998) rightly emphasise that it is important to focus
not on technical knowledge in group discussion, as this risks obscuring other aspects of
125
what they term ‗lay expertise‘. Similarly, as Michael (1996) explores, the reasons for
‗ignorance‘ about a new technology may be multiple. Consequently, providing information
risks homogenizing groups which, as described above, are selected to be heterogeneous.
However, to both ―promote and inform dialogue‖ (Armstrong et al. 2006) around technical
issues, particularly in the context of a technology of which there is especially little
awareness, it is often worth providing briefing materials.

I therefore wrote a booklet introducing molecular farming, which was piloted with the
group of undergraduate students, and on family and friends to gauge its readability. As can
be seen in Appendix I, the booklet contains a definition of molecular farming, proposed
advantages and disadvantages, crops used, types of containment, the institutions involved
in research and development, the extant regulatory frameworks and links to a website
containing further information12. Finally, a paragraph of information is provided on three
examples of molecular farming, HIV microbicides produced in maize, rabies antibodies
produced in tobacco and gastric lipase produced in maize. Two of the examples had
previously been used in studies of molecular farming (HIV by CWR&D 2005; gastric
lipase by Einsiedel and Medlock 2005), and allow some comparison between this and
previous work.

Building on the information booklet, the second meeting focused on perceptions of


molecular farming. This session was more actively moderated than the first, as I was
interested in exploring more specific questions. However, participants were still able to
spend the majority of the time exploring those facets of molecular farming that they felt
were particularly important or interesting. Participants were asked for initial responses to
the information provided, before talking in more specific terms about the examples
provided in the information booklet. The examples of an edible vaccine for hepatitis B
produced in bananas and insulin produced in safflower were also introduced in some
groups. The former prompted significant discussion, explored in detail in Chapter Five,
while the lack of familiarity with safflower oil resulted in little discussion. A number of
proposed means for containment including greenhouses, physical isolation and sterile

12
www.foodmedicinegenetics.org This address is no longer active, but the website can be accessed at www.rjmilne.com

126
plants were introduced13, followed by discussion of claims made about molecular farming
by scientists, environmental organisations and patients organisations. The relationship of
molecular farming to existing plant based medicines was then explored using a quotation
from the Biotechnology Industry Organisation. These prompts are reproduced in Appendix
III. Participants were finally asked to discuss whether molecular farming represented a
medical or agricultural technology. In Group F, I modified the protocol to introduce this
question at the beginning of the group meeting, to examine whether it made any difference
to group discussion; it did not appear to.

Between the first and second meetings of two of the groups (D and E) there was national
news coverage of molecular farming (Daily Mail and Daily Telegraph, 6th March 2007).
This provided further exposure to the subject and an opportunity to examine the effect of
news coverage on information. While two participants in Group D had been aware of the
coverage in the Daily Mail, only one had read the story, which led with the headline
‗Human Genes in Your Food‘. Rather than influencing the direction of discussion, it
seemed to act to reassure the group that participation in the research was worthwhile. In the
other group, a Daily Telegraph article about the transient production of pharmaceuticals
using a viral infection method had been picked up by two participants, but again did not
feature significantly in group discussion. While this highlights the relative paucity of media
coverage of biopharming, it is insufficient to make any major conclusions about the role of
such coverage in the formation of attitudes to biopharming.

A significant issue in the second group was my position as researcher. Having written the
information booklet on a topic that participants were unfamiliar with, at times I became
positioned as the ‗expert‘ in the room. While I was seeking to explore ways in which the
unfamiliar was rendered familiar through relating it to existing technologies and concerns,
it became clear that at times participants attempted to make the unfamiliar familiar by
simply asking me. As it did not overly hamper group discussion, this was not in itself a
problem. That participants seek to find out more information on which to base their
discussion when faced with questions about unfamiliar technologies, represents one way
through which they attempt to ‗place‘ biopharming. In addition, such questions are often
13
For a full list, see Appendix II
127
instructive about the ways in which participants approach biopharming, with queries along
the lines of ‗would they [plant made pharmaceuticals] be similar to a herbal thing‘ or
‗would they tell us if they were changing our medicines‘ revealing broader concerns and
understandings of pharmaceutical and herbal medicines. The group discussions were
recorded, transcribed and analysed, as I describe in section 3.4. Before that however, I
introduce the second strand of my research, which considers how public placings of
biopharming are paralleled in expert discussion.

3.2 Interviews and research documents

Focus group research aims to capture how publics relate biopharming to the narratives,
materials and places of existing medicines and foods. In addition, my research aims to
examine the resources and comparisons drawn on by biopharming researchers in
approaching the same questions. To do so, I make use of qualitative semi-structured
interviews with researchers. Such interviews have been used extensively within science
studies to examine the discourse of scientists, as well as their relationship with publics,
regulators and other scientists (Brown and Michael 2001; Burchell 2007; Kerr,
Cunningham-Burley, and Amos 1997).

In examining public and researcher discussion of biopharming ‗side by side‘, it might be


thought preferable to parallel the public focus group research by using a similar method to
reach experts (as for example Tutton 2007). However, such groups are both complicated to
convene (Cunningham-Burley et al. 1999) and, potentially disadvantageous. In particular,
while conducting focus groups within the confines of one laboratory might lead to freer
discussion, it would also involve imposing a group setting in a highly hierarchical
environment, exacerbating the hierarchic problems described in section 3.1.2. Constituting
more homogeneous groups be it of professors or post-docs from across laboratories would
be both less naturalistic and a significant logistical challenge.

Interviews provide information about events and topics and reveal the perspectives and
discursive practices of interviewees (Hammersley and Atkinson 1994). As such they
function as both a ‗resource‘, in the sense of producing ‗facts‘ about the social world and as
128
a ‗topic‘, as the discursive repertoires and rhetorical strategies through which interviewees
perform these facts are analysed (Byrne 2004). As do focus groups, they emphasise how
subjects frame and understand issues (Bryman 2001). They thus represent an eminently
suitable approach to examining how experts ‗place‘ biopharming.

The conduct of interviews within the social sciences ranges from "the highly structured
(akin to questionnaire survey in which the researcher asks pre-determined questions in a
specific order), through the semi-structured (where the researcher and participant[s] set
some broad parameters to a discussion), to the relatively unstructured (akin to a friendly
conversation with no pre-determined focus)." (Cook and Crang, 1995: 35). The interviews
in this research took the form of semi-structured ‗conversations with a purpose‘ (Burgess in
Byrne 2004), typically lasting around an hour. Adopting a semi-structured approach
ensures that there is some element of comparability between interview transcripts, while
allowing the expertise, interests and values of individual researchers to emerge.

Given the variation in the focus of each researcher and the evolution of both my
understanding of biopharming and of the project itself during the research period the
interview schedule varied in exact content from one to the next. As with the focus groups,
the aim of the interviews with scientists was to explore the meanings attributed to
biopharming and its relationship with existing agricultural and pharmaceutical
technologies. The interviews can be seen as ‗insider accounts‘ (Hammersley and Atkinson
1994) of biopharming, which describe the state of the technology, and provide another
layer to the accounts provided in review and research literature. Moreover, these accounts
are ―performances through which informants enact biographical, self-presentational and
explanatory work‖ (Atkinson 2005:9). As such, interviewees‘ responses represent a
particular performance of biopharming, through which it is placed in relation to agricultural
and pharmaceutical narratives, technologies and institutions. However, interviews also
involved descriptions of the materials of biopharming, often accompanied with images, and
in one case by a tour of the laboratories of biopharming research.

129
3.2.1 Selecting Interviewees

The first challenge in interviewing is in selecting who to interview. In attempting to


examine expert discussions of biopharming and enable some element of comparability with
their lay counterparts, it was necessary to identify relevant experts working within a similar
context to the focus group participants. Identifying European experts in biopharming was
simplified by the existence of the EU-funded ‗Pharma-Planta‘ project, launched in February
2004. Through contacting members of the consortium, and in combination with research
publications, I was able to capture a range of researcher perspectives on the development of
biopharming within the European Union.

The Pharma-Planta Constorium is an EU Framework Six Project whose primary goal is to


develop an approved production pipeline for plant made pharmaceuticals. Pharma-Planta‘s
aim is to develop an entire production chain by taking candidate pharmaceutical molecules
from the plant through to phase I human trials in Europe. The Pharma-Planta Consortium
(PPC) comprises 34 institutions including public institutes, SMEs and larger industrial
collaborators from 11 European Member States and South Africa (PPC 2007). These are
listed in Appendix II. As represented in Figure 3.3, the consortium is divided into six ‗work
packages‘ (WP), each of which focuses on a different aspect of the product development,
represented in a linear flow from bench to clinical trial.

The division of labour between the workpackages is described in PPC documentation thus:
(PPC 2007), WP1 provides the target molecules and the assays for their detection in
transgenic plant material. WP2 considers the potential environmental impact of
biopharming and leads interactions with appropriate regulatory agencies and other
stakeholders. WP2 also interacts with WP6 in the development of clinical trials dossiers.
This workpackage is the closest the PPC comes to considering the ethical, legal and social
implications (ELSI) of their research. WP3 provides expression platforms for the fast-track
molecules and generates the bulk material for their production. They select and develop the
plant lines that will express the recombinant antibodies, particularly focussing on maize and
tobacco. WP4 is the development loop, which includes a diverse range of expression
platforms and technologies not involved in the main trajectory of the consortium. WP5

130
oversees scaling, processing, quality assurance and quality control. It also leads interactions
with regulatory agencies concerned with pharmaceutical production. Finally, WP6 is
charged with organizing and performing clinical trials of the PPC fast-track molecules
(PPC 2007).

Figure 3.3 The structure of the Pharma-Planta Consortium

The organised and clearly defined nature of the PPC made it an ideal focus of research, and
answered the crucial question with which one starts interview research – who to interview
and how to contact them (Hammersley and Atkinson 1994). The consortium provided me
with a clearly defined research field, and identified the key researchers and groups involved
in biopharming research in Europe. This contrasts with others‘ difficulties in bounding the
research field in interviewing biotechnology researchers (Desmond 2004). Not only did the
management structure of the consortium allow me to clearly identify important
‗gatekeepers‘ and ‗obligatory passage points‘ within it, but the division of the consortium
into work packages ensured that I was able to target particular individuals or groups in the
aim of covering the whole consortium and thus, in the PPC‘s own terms, the development
of biopharming from target molecules to clinical trials.
131
Moreover, the representatives of workpackages two and five also provided useful insight
into relationship between the PPC and the development of European biopharming
regulation. The remit of the PPC also includes the elaboration and demonstration, or ‗road-
testing‘ (Sparrow et al. 2007), of a European regulatory pathway for biopharming. I
originally intended to supplement researcher interviews with a series of interviews with
representatives of the relevant regulatory bodies, particularly the European Medicines
Evaluation Agency (EMEA), experts from the European Food Safety Agency (EFSA) and
the competent authorities for the administration of regulation within the UK. In terms of
food and agriculture, this proved relatively simple as the majority of those involved in an
EFSA ‗self-tasking exercise‘ to develop regulation for biopharming are also members of
the PPC. In contrast, despite repeated requests, it was impossible to arrange an interview
with a representative of EMEA. Instead I was referred to the emerging regulatory
documents as the official line of the agency. However, as described by members of WP5,
consortium members themselves had trouble contacting pharmaceutical regulators without
having a product for them to assess.

3.2.2 Recruiting PPC Members

The PPC provided my research with a pool of experts with a particular vision for
biopharming, engaged in the development of both the technology and its regulation. The
project is split between six ‗workpackages‘ addressing different stages of a linear
translation from basic research to tested pharmaceutical product. The distribution of
research institutions between the workpackages is shown in Figure 3.4. Some institutions
have more than one group involved in the consortium, notably St George‘s Hospital,
London, the Fraunhofer Institute and RWTH University, Aachen, Germany and the John
Innes Centre, Norwich, UK. These represent important hubs of expertise in molecular
biology, plant biology and medical research. I visited all three of these central institutions
during the research and conducted interviews with researchers from each.

132
18 PPC researchers were contacted, of whom 12 agreed to be interviewed14. These 12
included researchers from each work package, thus tracking the technology along the linear
model presented above. Here the extent to which this model is a simplification, even for
those involved in the consortium, is immediately apparent, as many of the researchers
interviewed are members of more than one work package. For example those in WP4,
seemingly excluded from the main thread of the project, are often involved in the ‗core‘
project, although this is not always the case. The workgroup affiliation of the researchers
interviewed in the project is presented in Figure 3.5, in the rough order in which interviews
took place between October 2005 and October 2006. Professor L., a central participant in
the consortium was interviewed twice, both at the start of the research, to explore the
Pharma-Planta Consortium and establish contacts with other consortium researchers and in
March 2007 following the conclusion of the majority of the focus group stage of research.
Given that some researchers bridge more than one work package, the final distribution of
the interviews across the consortium is re-presented in figure 3.6.

At the time of research some PPC participants, particularly those involved in WP6, were
only nominally involved in Pharma-Planta, as their role is to test the final product in a
clinical trials setting. Of particular interest to me in examining the relations between
biopharming with agriculture and existing pharmaceutical production were workgroups two
and five. These are two of the explicitly ‗outward looking‘ projects, focused on finding a
‗natural home‘ for biopharming among agricultural and pharmaceutical regulation. I
discuss this latter role in more detail below.

As in Desmond‘s (2004) study of agricultural biotechnology researchers in Ireland, it


became clear that ―similar stories were told, discourses drawn upon and reports referenced‖
and that I was being ―asked had ‗X‘ or ‗Y‘ been interviewed, given their central roles in the
process‖. Given that participants were contacted on the basis of their involvement in a
particular project, it is perhaps unsurprising that at times I received the ‗project line‘.

14
Professor F., a leading US researcher in biopharming, was contacted during a visit to London, and a brief
interview undertaken. Due to the impromptu nature of the discussion, it was not possible to record the
discussion, although extensive notes taken informed the development of the research and of subsequent
interviews
133
Moreover, within the UK, and between the UK and Germany, there is a relatively small
biopharming community.

Figure 3.4 The Figure 3.5 Figure 3.6


distribution of Researchers
Distribution of
PPC institutions interviewed15
researchers
interviewed

WP1 9 Pseudonym WP No.


WP
interviewed

WP2 3 Professor A. 4,5,6 1 1

WP3 9 Professor B. 4 2 5

WP4 15 Professor C. 2 3 2

WP5 4 Professor D. 3 4 5

WP6 1 Dr E 2 5 2

Professor F (Non-PPC 6 2
member)

Dr G. 4

Dr H. 3

Dr. I 2, 5

Dr. J 5

Dr. K 4

Professor L. 1,2 ,3 ,6

Dr. M 2,4

15
Some institutions and individuals are involved in more than one workpackage
134
In particular, discussion of regulation, the public or of decision making in the PPC often
involved my being referred back to the same central participants, particularly Professors L.
and C. The latter is a biosafety expert, involved in debates around first generation
genetically modified crops, and provided an important introduction to the questions of
containment and the position of biopharming within regulatory structures. Professor L. on
the other hand is one of the pioneering figures in the field. Despite this emphasis on
particular experts there remain significant differences in researcher descriptions of
biopharming.

Of the other researchers contacted, Professor D is perhaps the most peripheral to the core
project of the PPC, and along with Professor A. is involved in the ‗development loop‘ for
future products. All except Drs H, J and K are UK researchers, representing a significant
proportion of the UK researchers involved in the consortium, while these latter were
interviewed at the Fraunhofer Institute, Aachen, a key site at which facilities for growing
and processing biopharmed materials are under development. The interviews took place in
a variety of settings, the majority in the office of the interviewee, while others were held in
the researcher‘s home or in cafés when they visited London.

While a dualistic distinction between ‗powerful elites‘ and ‗powerless others‘ is


problematic (Smith 2006), as a doctoral student interviewing researchers who often held
professorial positions within universities, there are obvious challenges involved in
"studying up" (Desmond 2004; Smith 2006). In the sense of defining elites as those with
‗elite knowledge‘, science studies has always been concerned with studying up, or at the
very least studying across (Latour 2005). As the purpose of the interviews was to explore
biopharming in more detail, and researchers‘ representations of its developments, this
relationship did not pose a significant problem.

Given my interdisciplinary background and institutional affiliation I made a significant


effort to engage with researchers within a ‗scientific vernacular‘ or ‗argot‘ (Bryman 2001).
My undergraduate training was in a mix of biological and social sciences, while my
research was funded by the interdisciplinary Institute of Human Genetics and Health at
UCL. I have thus been located within both a biology as well as social science department

135
throughout my thesis, and have spent time working in human genetics laboratories as well
as conducting social science research. I also attended an undergraduate course in plant
biology and familiarised myself with the research literature in biopharming prior to the
interview process. I felt this allowed me to probe in more depth into questions such as
glycosylation and containment (discussed in Chapters Five and Six), and consequently to
explore the materials as well as the discourses of biopharming. In addition, as Burchell
(2007) describes, engaging with researchers on their own terms contributes to a relaxed
atmosphere conducive to researchers fully elaborating their views on biopharming.

However, the positions of both interviewer and that of the interviewee are fluid through the
course of interviews. In my research, this was notable as discussion shifted from ‗technical‘
to ‗social‘ questions and from a narrow focus on the work of the interviewee to reflections
on the Pharma-Planta Consortium and biopharming as a whole. Rather than adopt a single
protocol for each interview, the questions asked were tailored to the relevant expertise of
the PPC member. However, all interviews started with an exploration of the scientist‘s own
biography and their route into the Pharma-Planta Consortium. A number of key topics
identified through the biopharming literature were also covered in each interview – the
challenge of containment; glycosylation and the equivalence of pharmaceutical proteins;
and their vision for the future development of the technology. In turn the answers to these
questions led to a re-reading and analysis of the research and review literature in
biopharming.

Understanding research accounts as social performances embedded within organizational


contexts, as social rather than individual narratives, emphasises the need to connect
interview data with the organization of the PPC consortium as a whole, and to situate the
commentary provided in interviews within that of the research field. To do this, I
supplement and combine the interview data with reviews of biopharming published by PPC
researchers during and shortly prior to the development of the consortium. The relationship
between reviews and interviews is recursive – review papers of the field were used to
identify potential informants, while in interviews, informants themselves frequently
referred me to back to further (or the same) reviews and research documents. In particular,
as I adopted the role of a relatively informed questioner with a background in both science
136
and social science, interested in exploring further the technical details of the technology,
such review documents represent an input into and natural extension of the interview
process. There is thus an interplay between written and oral accounts of biopharming.

3.2.3 Just Talk? Research Documents and Reviews as Narrative

Science circulates, according to Latour (1988), through ‗immutable mobiles‘, not least of
which are research papers and technical descriptions. Such primary research documents are
often accompanied on their journeys by a second form of scientific publication, that of the
review paper. If the first transmits the basic information of science, and the means of
reproducing knowledge in the Gabon as much as in Paris, the second provides the
motivating vision for this reproduction.

The interviews undertaken for the research represent a first site at which researcher
‗placings‘ and expectations of biopharming are explored. Review papers represent a
second, in which stories of the past and future of biopharming are laid out, and the
problematic relations with agriculture and pharmaceuticals explored. While the individual
research paper looks inwards towards the discipline, the purpose of the review is to look
outwards, to assess the landscape within which biopharming is emerging. As such, review
papers, in a similar, although importantly distinct, fashion to interviews, function as both
resource for exploring the actual status of biopharming, and topic, for considering how
biopharming is presented and performed.

Myers (1991) analyses two reviews of work in genetics, one by Francis Crick, the other by
James Darnell Jr. He presents the reviews as acts of story-telling, as acts of narrative, which
―draw the reader into the writer‘s view of what has happened, and by ordering the recent
past, suggests what can be done next‖ (1991: 46). Reviews represent a key site within the
scientific literature where expectations and promises are laid out, where the future of
technoscience is performed to shape the present in a persuasive fashion (see Chapter Two).
As such, review papers form a complement to interviews with researchers in that both
represent forums within which researchers expand ‗away from the data‘, albeit with caveats
about the inability to interrogate their arguments.

137
Reviews of biopharming were identified through literature searches and through the
Pharma-Planta website (www.pharma-planta.org), which gives a bibliography of recent
publications by consortium members. The reviews selected incorporate a range of authors,
(although some obviously predominate, particularly those from Fraunhofer IME in Aachen
and St George‘s in London, the institutional centres of the consortium), and stretch over a
period of time, including the period shortly before the consortium formed. Unlike the
interview process, this captures the evolution of biopharming during the history of the PPC.

The use of reviews from the Pharma-Planta consortium alone is again both practical and
pragmatic. It allows me to combine the literature effectively with the interviews with
researchers, to identify continuities between dialogue and published accounts, and more
interestingly, to highlight where there are discontinuities or where discussions present in
interviews become closed down. As the majority of the review papers are co-authored, they
(ostensibly) represent the outcome of discussions between members of the consortium that
it was not possible to witness.

In addition, research papers represent the means through which researchers describe their
interaction with the material world, in a parallel to focus groups‘ discussions of their food
and medicine consumption practices and choices. Descriptions of plant and pharmaceutical
biology in biopharming reviews re-emphasise the role of materialities in the ‗placing‘ of
biopharming, and prevent the thesis becoming overly concerned with disembodied
discourse.

As well as PPC review papers in academic journals, an edited collection of papers on


biopharming was also analysed for this research. The collection, published in 2004 and
entitled Molecular Farming was edited by two PPC members, Stefan Schillberg and Rainer
Fischer and includes a series of chapters outlining the current state of the technology at
roughly the launch of the consortium (Fischer and Schillberg 2004). These include
perspectives ranging from research on plant hosts, to the perspective of the pharmaceutical
industry on the technology, the relation with field trials of first generation genetically
modified crops and a discussion of risk and the precautionary principle. The chapters of the
collection move through the book from basic research and production platforms, through

138
trialling and biosafety issues, to commercialization and regulation. These research foci echo
the six workgroups of the PPC, also situated along this linear axis from research to clinic,
and culminating in two chapters considering the precautionary principle (Ammann 2004)
and a ‗top down‘ view from the pharmaceutical industry (Bischoff 2004).

While research papers add to the discussions of biopharming researchers, the chapters by
Ammann and Bischoff highlight the importance of existing pharmaceutical and agricultural
institutions and products in expert discussion of biopharming. To extend discussion of how
biopharming is ‗placed‘ among these as it develops, I also draw extensively on regulatory
documents, and on researchers‘ interpretations of them. In particular, I focus on how
regulatory documents represent an important stage for encounters between the visions and
promise presented in interviews and reviews, and the ‗regulatory everyday‘ of
pharmaceuticals and foods. As regulations were continually updated through the thesis, this
analysis was ongoing throughout the writing up stage, up until the release of the final
EMEA guidelines on biopharming in June 2008, and the release of the initial EFSA
documentation in April 2008.

The combination of review and regulatory documents with interviews allowed a more
detailed and informed mapping of the terrain of biopharming. In addition, it allowed me to
identify when researchers were sticking to a Pharma-Planta ‗line‘ in discussion, and where
alternate perspectives were emerging around the ‗placing‘ of biopharming in relation to
existing technologies. Together, these expert perspectives complement those expressed in
focus group discussion to provide a clear picture of biopharming‘s relations with foods and
medicines within both expert and lay narratives, places and artefacts.

3.3 Engagement between Experts and Publics: ELSI and the Media

The purpose of the research is not to bring about a direct encounter between expert and lay
discussions, but to place these perspectives alongside each other to examine the
development of biopharming within its socio-technical context. However, the development
of novel biotechnologies has frequently been characterised by such direct engagement and
deliberation. In this section I briefly highlight the absence of significant public engagement
139
work around biopharming in Europe, and the concomitant or contributing lack of
significant media interest in the technology.

There has been little or no significant public engagement work within the PPC. The
executive summary of the consortium‘s 2007 annual report suggests that the project
involves ―working with regulatory authorities within the EU as well as public groups to
ensure that the production systems are as safe and as acceptable as possible‖ (PPC 2007:3).
However, perhaps a more accurate picture is presented by the PPC website, which suggests
that by working with EU regulatory authorities the consortium will ―ensure safety and
acceptance‖ (http://www.pharma-planta.org/aboutus.htm; my emphasis). Public
involvement is limited to ‗acceptance‘, and there is no space for engagement on other
terms. As the PPC report of 2007 continues, communication and engagement with the
public is undertaken predominantly through the consortium‘s website. However, this also
provides no opportunity for interaction with the consortium‘s research, and is infrequently
updated.

The second possible arena for engagement between publics and researchers, although less
direct, is the media. The mass media are both an actor in, and arena for, discussion of the
development of new technologies (Bauer and Gutterling 2007). Bauer and Gutterling
(2007) suggest three conclusions about the role of the media in discussion of
biotechnology. The first of these is that news media has a limited but definite effect on
public perception, directing attention onto issues and setting an agenda rather than
providing the public with a 'conveyor belt' of information and opinion about biotechnology,
either negative or positive. Secondly, they conclude that media selection of issues is
relatively autonomous, based on newsworthiness rather than being driven by science input.
Finally, they conclude that media coverage of biotechnology has evolved over time,
characterised by hype in the early 1980s, with this positive tone declining until 1999, before
becoming more positive again. Media coverage of biotechnology peaks around the
coverage of the cloning of Dolly the sheep in 1997 and the introduction of Monsanto's
'RoundUp Ready' soya to Europe in 1996.

140
Bauer (2002) extends analysis of the media to consider the ‗cultivation‘ of a division
between red and green biotechnologies. In terms of the balance of media coverage, Bauer
(2002) has suggested that the media has cultivated more positive attitudes to medical than
agricultural biotechnology and has reinforced the division between red and green
applications. Bauer and Gutterling (2007) argue that this split took place following the
'watershed' events of Roundup Ready and Dolly. They state that since 1973 half of all
articles on biotechnology have concentrated on its medical potential, suggesting that this
reflects a 'general trend for medicalisation in this strand of news'

To examine effectively the ways in which molecular farming relates to existing applications
of medical and agricultural biotechnology, media coverage must therefore be considered
potentially influential. For two reasons, no deep analysis of media coverage has been
undertaken for the thesis. The first, pragmatic reason, is the logistical challenges of adding
to the large quantity of data already gathered. Secondly, media coverage of biopharming
has also been characterised by its paucity. There has been very little media coverage of
biopharming in the UK. The peak in coverage over the last ten years occurs around the
2004 launch of the Pharma-Planta Consortium (PPC) itself. Seven articles from this period
illustrate media discussion of biopharming. Four are taken from national newspapers in the
UK, two from regional newspapers and one from the website of the BBC. These pieces
were titled:

Britons to head up GM vaccines trial (Daily Mail July 13, 2004)

Medical crops coming soon: Human trials of GM drugs could be five years away (The
Guardian July 13, 2004)

GM Plants will be used to create new AIDS vaccine (The Independent July 13, 2004)

GM crops may be grown for vaccines (The Times July 13, 2004)

Vaccines grown in GM crops 'within five years' (The Evening Standard July 12, 2004)

GM Trials to change life down on the pharm (The Birmingham Post July 13, 2004)

EU funding for GM plant vaccines (bbc.co.uk July 12, 2004)


141
None of these articles was more than 800 words long, and many featured almost the same
quotes from Professors Julian Ma and Phil Dale of the PPC, based on the PPC‘s own press
release, along with critical comments from Claire Oxborrow of Friends of the Earth. The
description of the work of the consortium, as can be seen from the headlines above,
concentrates on the new use of agricultural biotechnologies. In the body of the texts,
Professor Ma acknowledges the ‗contentious‘ nature of the research resulting from the link
with GM, but counters this in the Independent with the argument that "the cost of doing
nothing is measured in millions of people who will die from preventable diseases". Medical
imperative is pitted directly against agricultural worry. Four of the articles also cite the role
of Phil Dale as leading efforts to isolate pharmaceutical crop production from conventional
farming. In contrast, the official press release accompanying the PPC does not mention
potential challenges arising from GM production. Given the sparse nature of media
coverage, its influence on specific thinking about biopharming is unlikely to be significant,
although the ‗cultivation‘ effect described by Bauer may be important as a background to
discussion.

As biopharming moves forward, particularly if products do come to market, one would


expect media coverage to increase accordingly, and for the number of references to media
coverage in focus groups to increase. However, in the three groups conducted before the
Daily Mail coverage of early 2007 described in section 3.1.4, and in the one group which
was conducted several weeks after this coverage, no reference to newspaper or other media
reporting of biopharming was made. This is not to infer that the media is of no importance
as a source of information, but to conclude that biopharming does not as yet feature heavily
in reportage. As an avenue for future research, a close analysis of the way in which
developing media reports of biopharming frame it in relation to agricultural and
pharmaceutical biotechnologies will make a useful addition to the current research.

3.4 Analysing The Data

Having established the means by which sufficient data can be gathered from a range of
sources, the analysis of this data becomes the central problem of research. Both groups and

142
interviews were recorded on MiniDisc using a multidirectional microphone. These were
then transferred to a PC, transformed into mp3 format and transcribed using ‗Express
Scribe‘ transcription software. This process was invaluable in managing the data generated
by the groups and interviews, and allowed me to become familiar with each individual
transcript. Indeed, transcription itself often forms the first stage of analysis (Bryman 2001).

Carey suggests that ―of all the aspects of using the focus group technique…the process of
analysis is the least agreed on and the least well developed‖ (1995:487). Nevertheless,
systematic analysis of group material is required to avoid imposing researcher‘s own
agenda on the findings (Bedford and Burgess 2001; Kitzinger and Barbour 1999).

Frankland and Bloor (1999) distinguish between two approaches to the analysis of
qualitative, particularly focus group data, that which draws on conversation analysis to
analyse the form of group discussion and a substantive approach to the content of
discussion. Given my interest in focus groups as a tool for examining the relational
positioning of biopharming, I adopt an approach aligned with the second approach, based
around what Seale (2004) terms ‗qualitative thematic analysis‘. This also brings together
the groups with the approach to the interviews described above, as both topic and resource.

In the first stage of analysis, the transcripts were read and re-read a number of times, and a
series of ‗index codes‘ attached to sections of data, becoming increasingly specific through
re-readings. I started out with paper copies of the transcripts. However, following the initial
coding stage, NUD*ist, a qualitative research program, was used to further code and assist
analysis of the data. Due to the unwieldiness of the NUD*ist software and departmental
expertise in the use of Atlas.ti, I re-entered and recoded the group transcripts in this latter
software. While concerns that analysis may simply be turned over to text-searching and
misguided attempts to automate an interpretive process (Frankland and Bloor 1999;
Waterton and Wynne 1999) may have some basis, the ability to move, manipulate and
rapidly retrieve large quantities of transcribed text and audio files has been invaluable in
this thesis.

This iterative ‗qualitative thematic analysis‘ (Seale 2004) led to a coding framework of
around over 100 initial codes. This first stage was followed by the elucidation of larger
143
themes which are approached in varying ways between different individuals and different
groups, but which are present across groups. Research interviews were similarly coded,
first separately and then alongside the focus group transcripts. The coding framework is
attached in Appendix IV.

While not involving the quantification of qualitative data, examining how themes are
repeated through the process of coding prevents ‗impressionistic assumptions‘ about the
data (Kitzinger and Barbour, 1999: 17). During the research process, both lay and expert,
the transcripts were constantly revisited, and new approaches to their interpretation taken.
This process continued into the initial drafting of the empirical chapters of the thesis: as
Burgess describes, ―insights continue to develop through the process of drafting as well as
these earlier stages‖ (1996:133).

Focus groups represent an arena for discussion, debate and elaboration of positions through
conversation. Analysis and presentation of data must therefore endeavour to capture this
process. Kitzinger (1994) particularly emphasises the importance of attending to interaction
in group discussions, rather than separating comments out as individual ‗attitudes‘.
Consequently, research such as that by Kerr et al. (1998) employs extended quotations.
Throughout the thesis I have attempted to ensure that focus group discussions are
contextualised and that their development through interaction can be observed.

In developing the empirical chapters, focus group discussion was combined with the
transcripts from interviews with biopharming researchers and a reading of biopharming
papers, particularly review papers. These were transcribed and coded in much the same
way as the focus group data. Focus group and interview data were coded within the same
‗project‘ in Atlas.ti, which allowed the same codes to flow across both sets of data, but also
for the separation of the transcripts into ‗families‘ such that either could be examined apart
from the other.

In this chapter, I have introduced the methods through which I have collected and analysed
lay and expert accounts of the place of biopharming among everyday technologies and
practices of food and medicine. My aim in doing so was to examine how the promises,
origins, materials and spaces of food and medicine are transformed by biopharming and
144
how the novel biotechnology is itself transformed in the process. In the following chapter I
start to explore this co-production and placing through a discussion of lay and expert
narratives of biopharming.

145
CHAPTER 4: FAMILIAR NARRATIONS AND
PLACED EXPECTATIONS

Biopharming as a human pharmaceutical production technology remains a prospect. No


human pharmaceutical product and only one veterinary medicine have yet made it through
clinical trials. As such, its future relies on expectations of the entry of products into clinical
trials and the extrapolation of existing research successes. However, it is not an unusual
example. Biotechnologies commonly operate within ‗regimes of hope‘ constituted through
the circulation of promises, expectations and hopes. The promises, expectations and pasts
of biopharming presented by researchers represent the first point of encounter between
publics and experts. These encounters allow social and financial support for novel
innovations to be mobilised among ‗communities of promise‘ which form around
anticipations of future success (Martin, Brown and Kraft, 2008).

Imagined technological futures represent a key part of the repertoire through which
biotechnologies are ‗placed‘. However, focussing on the future alone is asymmetrical.
Drawing on the work of Edward Said I have suggested that the authority of expectations
derives from association with that of parallel ventures. Moreover, focusing on the temporal
aspects of future narratives obscures their spatial content. In this chapter I explore how a
symmetrical approach to the pasts, futures, times and spaces of biopharming contributes to
understanding how it is placed in relation to agricultural and/or pharmaceutical narratives.

I first consider how biopharming is placed through ‗familiar narrations‘, by association


with the experience and authority of existing pharmaceutical and agricultural technologies.
I start with discussion of expert narratives, as it is from these that the promise of
biopharming emerges. I then consider how discussion of biopharming in focus groups
relates it to the pasts of pharmaceuticals and foods. In the second part of the discussion I
describe how expectations of biopharming are ‗placed‘, both temporally as developments
from these pasts, and in ‗imagined geographies‘ (Said 1995). I describe how for both
experts and publics, the relation between the places of production and application of
biopharmaceuticals become central to the establishment of its promise.

Part I Familiar Narrations

4.1 Experts: From Agriculture to Biopharmaceuticals

Possible futures of biopharming are enabled through association with the authority of
parallel ventures. This is accomplished through placing the new technology within
narratives of agriculture and pharmaceuticals16. The interplay between public and expert
narrations of biopharming establishes the grounding for the formation of ‗imagined
communities‘ of promise. I first consider how biopharming researchers draw on agricultural
and pharmaceutical pasts to underpin the potential of plant made pharmaceuticals.

4.1.1 Narrative One: From Agriculture to Food

Agriculture is at the heart of the pharmaceutical promise of biopharming. In an interview


with a PPC researcher the development of biopharming is introduced as a question of plant
biotechnology, defined not through products, nor its process, but through the biographies of
PPC researchers themselves and of the technological platform:

Well the platform technology is a plant one isn't it, and all the technical hurdles are
plant ones, you know, anybody can choose a molecule, the question is how do you get
it into a plant, how do you get it to express well in plants, how do you extract it from
a plant and purify it. And you know a medic, or an immunologist, aren't going to be
interested in that. All they're interested in is you know give me the final molecule and
I'll test it for you
(Professor L.)

The knowledges and expertise of the plant science and agricultural tradition are integral to
the future development of the technology. As the production of pharmaceuticals in

16
Throughout the empirical discussion in the thesis, I refer to ‗conventional‘ pharmaceutical production. By
this I mean industrial, processed or ‗mainstream‘ pharmaceuticals, as distinguished from ‗traditional‘
medicines, which includes herbal and alternative remedies.
147
genetically modified crops, biopharming emerges from the institutions of, and uses many of
the same technologies as, agricultural biotechnology. However, its links with agriculture
extend beyond the biotechnological.

Within the biopharming community, and even within the PPC, the agricultural form of the
technology remains fluid, and there is significant diversity in the range of crops adopted for
pharmaceutical production. One recent review (Basaran and Rodríguez-Cerezo 2008)
identifies 11 different crop platforms for the production of 40 plant made pharmaceuticals
currently in clinical trials worldwide, while another review lists 25 species which have been
used in research (Ramessar, Sabalza et al., 2008). Among these, the key distinction is that
between food and non-food crops. The selection of either as a production platform involves
a significantly different ‗placing‘ of biopharming in relation to existing agricultural
practice. Of the 11 crops identified by Basaran and Rodriguez-Cerezo, five are food crops
(maize, rice, potato, melon and spinach), while six are non-food (tobacco, plant-cell
cultures, lemna, safflower, Arabidopsis and canola).

Food and Experience

Early PPC work presented a dual research programme focused on maize and tobacco to
develop experience with food and non-food crops. By 2008 however, maize emerged as the
primary production platform for the PPC (Sparrow et al. 2007). Ramessar and colleagues
set out its advantages:

―Maize is one of mankind‘s earliest innovations… In the context of molecular


pharming, maize is advantageous because it has a well-established agricultural
production infrastructure allowing production to be scaled up and down rapidly in
response to demand‖
(Ramessar, Sabalza, et al. 2008:411)

Maize has benefits for PMP production in terms of ―familiarity, economics, technical
know-how, germplasm resources, a wealth of genetic knowledge, and a well-developed
agricultural infrastructure‖ (Sparrow et al. 2007: 5). The importance of familiarity and
technical know-how places biopharming in relation to agricultural practices, but also
locates its future in countries in which maize growing infrastructure exists. The established

148
agricultural infrastructure associated with maize accompanied by an established research
base.

PPC researchers are more knowledgeable about food crops in general and maize in
particular, as introduced in an interview with Professor C.. Here he places maize research
within the tradition of production and research involved in the development of a range of
desirable product qualities.

You get by far the best yields from a crop that has been bred for food ... You know
people talk about well, why don't we just find another plant that's not used for food
and domesticate that? But domestication takes hundreds and hundreds of years, and
to breed in all the characters you needed, including high yield, ability of seeds to
shed under threshing and all of that, it is not a trivial job.
(Professor C)

The expertise researchers have in particular plant species contributes to driving the choice
of technological platform. The reasons for choosing a food crop are that they are better
characterised, because they are a food crop. Scientific knowledge and infrastructure has
clustered around these crops. As such the choice of maize represents the self-reinforcing
weight of tradition –technological ‗lock-in‘ (Stirling 2008) generated by the co-production
of scientific knowledge and agricultural infrastructure. The agricultural and food
characteristics of maize, and most importantly the tradition of use, underpin its promise as a
biopharming production platform, as described in further researcher interviews:

I mean it‘s one of those awkward things because, in some ways, obviously people
choose edible plants because they're not toxic, they have no toxins, and an awful lot
is known about processing, because they're actually used.
(Professor A., emphasis added)

Although it is a potentially ‗awkward‘ point for researchers, the primary feature of food
crops is their relation to existing knowledge and expertise and the legacy of agricultural
research and production. The association with a tradition of agriculture and consumption
allows researchers to argue that the benefits of the use of food crops outweigh possible
risks. The use of well-characterised crops allows the PPC ―to dispel the misconception that
it is better to use less known and non-domesticated plants…instead of a well understood

149
and familiar species‖ (Ramessar, Sabalza et al. 2008: 410). At the forefront of the
advantages of maize are its relation to existing practices and knowledge, in the form of
infrastructure and existing consumption, and through the pinning of biopharming to the
success of humanity‘s ‗earliest innovations‘.

Experience and Assessment of Risk

The choice of food crops raises an obvious concern about the mingling of food and
pharmaceuticals. This is expanded in the 2008 guidance on PMPs from the European Food
Safety Authority (EFSA), which describes how:

―…some of these plants may be modified to produce novel compounds which are
biologically active in humans, in animals or in other non-target organisms, and are
not intended for food or feed use, nor to be widely distributed in the environment. The
potential risks of such plants will vary…‖
(EFSA, 2008: 10)

The familiarity and harmlessness of maize as a pharmaceutical production system contrasts


with concerns about the potential for the contamination of the food supply with medicines.
For some researchers though, the use of food crops also makes the risks of such
contamination easier to assess.

Ramessar, Capell and Christou (2008) suggest that this experience with the processing and
consumption of food crops also allows the risk of molecular farming to be effectively
assessed. Again, they argue that despite the fact that pharmaceutical crops may not be
intended for direct consumption, the tradition of such use allows the risk of inadvertent
consumption to be assessed:

―Cereals are advantageous … because they are food crops with FDA GRAS status,
having been consumed for millennia by billions of people. A sea of information exists
concerning the allergenic potential of cereals and dietary exposure studies, providing
easily accessible equivalence data for risk assessment comparisons.‖
(Ramessar, Capell and Christou 2008: 124)

Not only then is food, and its tradition of consumption, important in terms of research, but
the history of safe consumption is important in assessing the risks of consuming a

150
pharmaceutical containing variant. In addition, biopharming is discursively distanced from
agricultural traditions and geographies. Researchers point to differing located traditions of
maize agriculture to separate research infrastructure and expertise from the food
infrastructure required for production:

A food in one country is not necessarily a food in another country. So in the UK we


don't really grow maize as food, the conditions aren't right, you know, we grow
wheat, barley whatever. You know, you'll occasionally come across a maize field that
is grown for animal feed. We don't grow maize basically. So I don't regard maize in
England as a food product. So why shouldn't we grow pharmaceutical maize?
(Professor L.)

The problems of maize production are situated in particular cultural geographies of


consumption, which distinguish them from an imaginary of British or English food
production. Both agricultural production and research knowledge are situated in the above
extracts. However, researcher knowledge deriving from maize‘s food status is not co-
located with maize production for food.

Researchers establish biopharming as continuous with food applications of agricultural


research. They emphasise the importance of plants as a platform over pharmaceuticals as
products, and suggest that the best characterised plants, food crops, offer the best platform.
Consequently, the choice of food crop platforms is directed by traditions of food use which
have led to their use in recombinant biotechnology and farming. The experience and
expertise derived from these agricultural traditions shapes the form taken by biopharming
in the Pharma-Planta Consortium. The pasts of the technology shape its present and
constrain its possible futures.

4.1.2 Narrative Two: From Biotechnology to Botany

While biopharming is placed in terms of a lineage of agricultural research, it is also


associated with a tradition of pharmaceutical production. Two approaches to its
pharmaceutical antecedents can be identified in review papers and researcher interviews
which situate it in relation to the ‗extractive‘ and ‗biological‘ heuristics of the
pharmaceutical tradition. The early development of pharmaceuticals relied heavily on plant

151
extracts such as aspirin, quinine or digitalis, as well as complex biological products such as
anti-sera. The emergence of recombinant DNA technologies in the 1970s represented the
rebirth of this biological model of drug discovery and manufacture. It is this latter tradition
which I discuss first, as biopharming is placed within a narrow chronology of recombinant
biopharmaceutical production. This can be seen in an extract from a review of biopharming
by Gomord et al.:

―For several decades, genetically modified organisms (GMOs) have been used to
produce therapeutic molecules in a recombinant form. For a long time, the only
GMOs used for such production were Escherichia coli, yeast and mammalian cells.
Over the last 20 years, several proteins of pharmaceutical interest have also been
produced in transgenic plants‖
(Gomord et al. 2004:83)

In these terms, biopharming is clearly a ‗third wave‘ (Hopkins et al. 2007) pharmaceutical
biotechnology. It represents a novel approach to biopharmaceutical manufacture, but one
which is continuous with alternative recombinant systems. However, these biotechnological
lineages often emerge alongside the association of the technology with a significantly
longer history which extends the narrative back to the physic and botanical gardens of the
19th century and before. For example, in their introduction to Molecular Farming, Fischer
and Schillberg draw on a long lineage:

―Mankind has used plants as a source of raw materials and medicines for thousands
of years. From the earliest stages of civilization, plant extracts have been used to
obtain technical materials and drugs to ease suffering and cure disease.‖
(Fischer and Schillberg 2004)

The genealogy expressed above, in which biopharming is presented as a development from


existing pharmaceutical biotechnologies, is expanded and extended. The technology is
associated with innovation not in pharmaceutical biotechnology, but in the medicinal use of
plants themselves, a re-emergence of the extractive as well as an extension of the biological
heuristic of drug development.

The association of biopharming with plant extracts is part of the rhetorical opening of
papers which situates the narrative that follows. As such it is part of the strategy through

152
which the writer of a review shapes the literature of a field into a story to enlist the support
of readers to continue that story (Myers 1991; Law 2002). The narrative history into which
these brief paragraphs insert biopharming places it temporally, as continuous with
medicinal uses of plants. Mention of use of plants for medicine by the ‗earliest
civilisations‘ determines the salient historical nodes (Brown 1998) of biopharming, from
which its future development can progress. This point is made explicitly by Fischer and
Emans in a 2000 review of biopharming:

―We foresee that molecular farming will provide a basket full of novel medicines for
the diseases of the 21st century, just as plants were the source of medicines for the
Egyptians 3600 years ago‖
(Fischer and Emans 2000:295)

The pasts of botanical medicine provide the basis for imagining a revolution on a scale not
seen for over three millennia. Hence, although the biotechnological potential of
biopharming is revolutionary, these claims are authorised by association with the authority
of a ‗parallel venture‘ (Said 1975). Continuity between recombinant plant made
pharmaceuticals and traditional plant derived medicines is considered in the extract below
from an interview with a senior plant biologist from the PPC:

You know traditionally you think of plants making pharmaceuticals and you tend to
think of it maybe as rather alternative therapy, but actually that‘s not true, because
things like vinblastine, you know the cancer treatment are still grown in
periwinkles...So it‘s not, it‘s not such a weird idea as it sometimes seems, you know,
we're going to grow all sorts of vaccines in plants, that sounds really odd, but
actually if you're unfortunate enough to need treatment with vinblastine, that‘s
exactly where it would come from
(Professor A)

The less than everyday example of vinblastine is used to place molecular farming in a
continuum of plant based medicine, while distinguishing it from ‗alternative‘ medicines.
Moreover, the use of the contemporary example of vinblastine emphasises the continued
potential of extractive medicine. In this description, biopharming is both familiarised and
validated - it is equivalent to existing medicines and shares their potential.

153
However, the link with an extractive genealogy in the papers and interviews introduced
above introduces a relationship between PMPs and other extractive medicines, particularly
those currently marketed as ‗herbal‘. Despite the common link with a botanical tradition, an
explicit link with existing herbal medicines is rarely made in review papers. This
relationship is explored in interviews with PPC scientists:

RM: The other thing I suppose is the tradition of medicines from plants - is there
linkage there?
Yes, very, there is a very logical link, and I think there is also a lot of experience we
should be building on and we could be building on, and also public acceptance might
be largely influenced by this, because we know all those herbal remedies and so it
might be very easy to have someone accept pharmaceuticals coming from plants. So I
think yes…but using plants for pharmaceutical production we are underlying all
those strict regulations for biotechnology sort of for biotech products, for biologicals,
biologics. And so there are a lot of things we would be much better off dealing with
the guidelines for herbal remedies. A lot of regulations there would fit so much better
… We have the problem that we are always compared to frozen CHO cells…these
systems are not very similar at all. It‘s very difficult to try and 'be a CHO cell' with
our plants, while it would be so much easier to comply with the regulations for herbal
remedies.
(Dr H.)

In this interview, the researcher outlines multiple reasons for the link between biopharming
and the herbal tradition. She introduces both discursive and material advantages. Firstly,
framing biopharming in terms of herbal medicines establishes it as familiar and acceptable
to publics. As such, it serves as an argumentative device, as in the reviews. Secondly
however, she argues that this is more than narrative, and that the material of biopharming
itself is best understood in terms of herbal medicines as opposed to that of
biopharmaceutical production. The narration of biopharming purely in terms of its
pharmaceutical products is complemented by a narrative concentrating on production. This
focus on production is explored in a second interview with a PPC member involved in the
development of regulatory advice for biopharming:

I think we need to remember that plants are traditional producers of medicines. I


think one can only take that argument so far because people know that you get

154
certain medicines from a poppy and certain other things, and so there has been an
agricultural system developed around those in a particular way, whereas if you
introduce another gene into, or a medicine gene into a crop then, one would need to
develop a whole new kind of set of protocols for that crop … I think all the experience
with traditional medicine is quite useful in some respects, but in terms of the
familiarity and unfamiliarity then it‘s not so relevant.
RM: Are regulations around herbal medicines useful?
Yes, I think they are quite useful, er but compared with modern PMPs the regulation's
quite, quite lax, but it‘s the old problem, things which are traditional are natural and
wholesome and whatever, even if it's not really, it‘s the perception.
(Professor C.)

This extract starts with a reiteration of the rhetoric of the review papers introduced above,
positioning biopharming as not inherently novel in concept. However, the researcher moves
to distinguish traditional medicines from biopharming on the same basis used in the
previous interview to associate them – their context of production. He argues that
traditional botanical medicines can only be understood in terms of their production, and that
the act of genetic modification distinguishes biopharming from this process. Thus, the
association of biopharming with existing food agriculture is accompanied by a distancing
from the traditional knowledges and infrastructures of pharmaceutical agriculture. For
others within the PPC the problems of discursive association with herbal remedies
outweigh the potential advantages of the experiences described by Professor C. and Dr H.
This is explored further in an interview with a senior member of the PPC involved in the
development of pharmaceutical products:

RM: Is there a link with existing plant medicines?


Well yes and no. I think plants have always been a source of medicines, but the
industry have always been very quick to move over to chemical synthesis, and there
have really only been a few cases where that hasn't worked … And the reason why
there's not been a huge interface is because I think traditional plant medicines either
go that breeding route and so there's no genetic modification at all, so there's none of
those sort of safety overview that we have to think about, but also very few of those
drugs actually go through MHRA/EMEA kind of regulatory guidelines. Most of those
are kind of herbal medicines and they don't, they're not really regulated very well, so
I would never take a herbal medicine actually...so almost by definition we don't

155
follow their line. Also, I don't want to be known as a herbal medicine, because I
don't want that stigma of not being regulated"
(Professor L.; my emphasis)

In this interview, the association of biopharming with herbal medicines is by no means


obvious and is seen as something to be avoided. Unlike the previous two interviews, the
association of biopharming with a history of farming herbal medicines is ruled out entirely.
Instead, this researcher draws on a shorter history to argue that where possible,
pharmaceutical production has always moved away from plants. He thus challenges the
material associations between what I term ‗traditional‘ and ‗conventional‘ medicines.
Doing so enables him to make the argument that association with herbal medicines is
potentially deleterious to the development of biopharming. Rather than representing a
positive association then, a connection with herbals and their unproven potential risks
stigmatising the technology.

Professor L. establishes the achievement of approval by pharmaceutical regulators as the


goal of biopharming, and as the point at which equivalence between biopharming and
pharmaceutical biotechnology will be established. His position is reflected in the
development of review papers‘ narratives of biopharming as they attempt to strike a
balance between continuity and novelty. Continuity with plant based medicines is
complicated as review papers expand their narratives to emphasise the novelty of
biopharming in biopharmaceutical terms:

Plants have provided humans with useful molecules for many centuries, but only in
the past 20 years has it become possible to use plants for the production of specific
heterologous proteins
(Ma et al. 2003:794)

Plants have been used in medicine for many centuries but it is now possible to exploit
plants as bioreactors for the production of human therapeutic protein.
(Daniell, Carmona-Sanchez, and Burns 2004:113)

Plants and their products have been used for centuries to prevent and cure disease.
More than a quarter of all the medicines used in the world today contain ingredients
derived from plants. However, it is only recently that biotechnology has been used to
156
generate plants that produce specific therapeutic proteins, products that are
traditionally synthesized using recombinant microbes or transformed mammalian
cells
(Ma, Barros et al., 2005: 580)

In these extracts, the biotechnological history described at the start of this section re-
emerges. For experts the relation with plant based medicines is shown to be more complex
than might be suggested by the continuous use of ‗deep‘ genealogical introductions to
biopharming in review papers. Instead, biopharming represents a dual revolution, both in
the development of recombinant pharmaceuticals and in the use of plants. The authority for
the pharmaceutical promise described in the remainder of these reviews is provided by an
association with biotechnological and botanical traditions, but also a distancing from each.

There is no simple distinction between continuity and discontinuity in the association of


biopharming with existing pharmaceutical applications. Instead both are integral to
narratives of biopharming‘s development. Equally, the histories which are available are
approached in multiple ways. Hence the relationship between plant made pharmaceuticals
and botanical or herbal medicines is for some a positive and logical link, while for others it
is perceived as distancing biopharming from conventional pharmaceutical production.

Biopharming emerges from plant biotechnology, and traditions of agricultural research


shape the decisions made around the production platform. Here PPC researchers move from
considering this platform to concentrate on the status of the products. The relationship
between biopharming and previous uses of plants for the production of medicines is
contested. Long histories of plant based medicine are used to familiarise biopharming, but
the applicability of this history is contested, both in practical terms and in terms of the
desirability of a link with herbal medicines. Instead, researchers move to distinguish
biopharming from traditional medicine and re-align it as an alternative to conventional
biopharmaceutical manufacture.

For PPC researchers, the narrative placing of the technology involves a complex
arrangement of continuities and discontinuities, of novelty and tradition. A first
agricultural/plant based narrative establishes food crops as a logical choice for

157
pharmaceutical production. Secondly, a pharmaceutical narrative emphasises the role of the
biopharmaceutical product in distinguishing biopharming from traditional plant based
medicines.

4.2 Publics: The Serendipitous Pasts of Biomedicine

Interviews with researchers and analysis of research documents were accompanied by a


series of six repeated focus group discussions with members of the public in London and
rural England. The second group meeting focussed specifically on molecular
farming/biopharming, while the first meeting discussed food and medicine more widely to
allow the ways in which biopharming is ‗placed‘ to be analysed.

For publics, the relation of the novel technology with past events and experiences is
entirely dominated by medicines. Perhaps one of the most significant findings of the
research is that biopharming is rarely associated with the pasts of agricultural
biotechnology by members of the public. Where this tradition is raised, it is obscured by the
promising biomedical futures of the technology.

In general, focus group discussion of biopharming was positive, with participants seeing it
as a worthwhile application of pharmaceutical research. For many focus group participants,
seeing molecular farming as simply another form of drug production contributed to
familiarity and support. The benefit of placing molecular farming alongside conventional
pharmaceutical production is demonstrated in Group A (young urban male):

Euan: We trust normal drugs don't we, so why wouldn't we trust these if they're made
by the same companies, same thing really isn't it
James: They should have gone through all the right testing procedures...I mean that
whole Vioxin thing, didn‘t it, wasn‘t it Vioxin? Didn‘t it turn out they hadn‘t done the
monkey trials properly or something?
Anil: They skipped something because it didn‘t work properly or something
Rob: Well we assume that drugs that are produced from bacteria are all, I mean,
that‘s a living organism, it has the same potential as these plants do for perhaps
errors to occur, in the sense that animals could eat the bacteria
(Group A:2)

158
Group A are comparatively knowledgeable about current pharmaceutical production, and
are the only group to raise the question of the safety of current biopharmaceutical
production methods. However, the existing risks are used to justify the potential ones posed
by biopharming. While this group was positive about molecular farming overall, discussion
of the similarity between molecular farming and conventional pharmaceutical production
produced a number of worries, and introduces the idea that associations with other medical
trajectories are not without complication. A number of groups used the example of
‗Vioxx‘17 to highlight the potential weaknesses of the regulatory system and to challenge
the idea that a shared origin of molecular farming and conventional medicine should result
in more trust.

However, other examples were drawn on more commonly than Vioxx in framing
discussion of biopharming. In particular, discussion often links biopharming research to
past successes in medicine, most notably to vaccination and the discovery of penicillin. The
latter is the most prominent event through which expectations of molecular farming are
placed by focus group participants.

Penicillin was raised by four of the six groups as an example of scientific success when
discussing molecular farming and plays an important role in authorising the hopeful
narratives discussed in section 4.4. A similar role is played by vaccination, which is seen as
a significant success for medical research and progress in group discussions. Both of these
can be seen as discussion in Group E (older rural mixed) develops between the first and
second group meetings:

Douglas: I think it‘s interesting though, some of these things if you don‘t actually go
into it and do the research in it and move along, then you‘re just going to stick where
you are, so you‘ve got to be looking at stem cells, you‘ve got to be doing these things,
otherwise you‘re going to be dead in the water sooner or later
Jane: It‘s the next step; it‘s a natural next step to take. If people had taken that view,
we wouldn‘t have any vaccines

17
Merck introduced Rofecoxib (Vioxx) in 1999 as a treatment for pain associated with osteoarthritis. Despite
evidence that it might contribute to cardiovascular disease, none of the studies included in Merck‘s
application to the US Food and Drug Administration evaluated cardiovascular risk. Although it was later
found to increase the risk of cardiovascular disease and was withdrawn from the market, Merck faced a large
number of law suits from affected individuals (Krumholz et al. 2007).
159
(Group E, meeting 1)

In the second meeting, this discussion was picked up by Paul:

Paul: Aye, but I mean if you look at it another way, if somebody said oh well, here‘s
some mouldy cheese and that‘s going to cure all your ailments, then you would‘ve
pooh-poohed them whatever and would have never have got off the ground, and…
RM: You‘re talking about penicillin?
Paul: Yeah, which is basically what it was in the first place.
(Group E, meeting 2)

In these extracts progress, both intentional and serendipitous, is at the heart of the medical
imaginary which underpins support for biopharming and medical research in general.
Medical innovation is seen as a ‗natural next step‘ but, as highlighted by the example of
penicillin, one whose direction is unpredictable. Descriptions of both vaccines and
penicillin describe them in terms of medical research, and its ability to offer progress. The
ability of medical science to capitalise on the serendipitous discovery of the natural
qualities of penicillin represents an important and widely known story of research success
to focus group participants (cf. Merton and Barber 2004). For the group of older rural
women (D), this success provides reason to trust the scientists developing molecular
farming:

Molly: Yes, and I was trying to think, penicillin, that was natural wasn't it, mould, so
I was thinking some of these drugs might have come from natural…
Sofia: Antibiotics as well you see, like the antibodies
Jean: It‘s just what they add to it
Molly: These scientists, they're very clever people. I can't remember, because I don't
know anything about it, but I remember them saying at school that it came from
mould, penicillin
Sofia: I do
Molly: That‘s been a help to mankind hasn‘t it, so all the drugs that we use aren‘t
always bad, they‘re not all from…
(Group D, meeting 2)

This extract combines the history of medical developments with the intentions and
characteristics of those performing the research. Since the Vannevar Bush‘s ‗Endless
Frontier‘, penicillin has served as a prime exemplar of scientific progress (Bush 1945). Bud
160
(2007) describes how it became a metaphor for the triumph of modern medicine in the
1960s, and this persists in its use as an anchor for framing discussion of medical research.
These narratives draw on the power of penicillin and vaccination as breakthroughs or
‗commemorative moments‘ (Brown, 2000) representing great leaps forward in a
metanarrative of progress through innovation (Felt et al. 2007). However, the true
authorising power of such narratives of medical promise derives from their unpredictability,
offering an interesting contrast with the adoption of the precautionary principle for first
generation agricultural biotechnologies18. In place of avoiding unknown harms on the basis
of scientific uncertainty, arguments based on penicillin imply that medical research should
continue in search of unknown benefits.

The insertion of molecular farming into a narrative of medical progress is expanded in the
extract from Group C below. While group C (older urban mixed) was generally the most
sceptical of the research groups, Peter continually argued for more support for molecular
farming.

Peter: …I think there‘s been lots of progress in terms of health and so on in past
centuries, which because, in a sense people made tests, people explored the unknown.
I mean I remember vaccines when I was a boy, and I guess before that, people had
died of things which you can live with. I don‘t really go too far with this sort of
natural and unnatural, I think we talked about that last week, I think that plague is
natural in that sense.
(Group C, meeting 2)

Stories of past success authorise future medical research. Peter also uses reference to the
tradition of progress and research in medicine to dispute the importance of ‗nature‘ in
judging new biotechnologies. In researcher imaginaries of biopharming, herbal medicines
situate its potential, as a useful source of knowledge about the practices of pharmaceutical
production in plants. At the same time they try to avoid linking PMPs with a framing of
herbal medicines as unregulated and ineffective. They describe with exasperation irrational

18
Von Schomberg offers a definition of the precautionary principle as follows: "Where, following an
assessment of available scientific information, there are reasonable grounds for concern for the possibility of
adverse effects but scientific uncertainty persists, provisional risk management measures … may be adopted,
pending further scientific information for a more comprehensive risk assessment, without having to wait until
the reality and seriousness of those adverse effects become fully apparent" (2006:37)
161
support for ‗natural‘ remedies. Here Peter adopts a similar approach to the valorisation of
the natural, opening up the multiple interpretations of the ‗natural‘. The association of
biopharming with a tradition of experimentation is more important than that with natural
medicines. As the discussion develops, Peter expands his point:

Peter: But it comes back to this thing, I‘m not sure you can naturally assume that a
potato that has been molecularly farmed is automatically the same thing as kind of
ancient plants. My reasons for being open to molecular farming are more to do with
the good it could do rather than making a link between this is good and wholesome
and natural
(Group C, meeting 2)

Peter argues that the positive medical potential of the technology outweighs considerations
of other characteristics, essentially suggesting that its relation with traditional medicines is
irrelevant given the pharmaceutical promise of the technology. At the same time, he links
biopharming with the biomedical narrative introduced earlier. For other groups,
establishing the relation between biopharming and the herbal tradition was more complex.
All groups were asked whether they saw molecular farming as related to the tradition of
herbal medicines. While a number of groups saw a link to plant derived medicines in
general, none were convinced that molecular farming represented a similar use of plants to
herbal remedies, as highlighted in the following extract from Group F (young rural mixed)

Katie: Because they've been put there, haven't they. They're not like herbal remedies
Alex: It‘s making a plant grow in a certain way isn't it, to produce it

RM: So it‘s not like herbal medicines?
Rebecca: No, I wouldn't see it as a herbal medicine. Not even if it was in a banana
and I was eating the banana, I'd still think it was a chemical something
Alex: Yeah, it‘s still the result of a chemical reaction, it has just not taken place in a
vat, it has taken place in something green and growing
(Group F, meeting 2)

In this discussion, a distinction is made between molecular farming and herbal remedies on
the basis of the form of production. Rebecca and Alex describe the products of
biopharming as continuous with those of chemical pharmaceutical manufacture, despite the
plant-based nature of production. As do researchers, focus groups attempt to place
162
biopharming in relation to herbals, and consider the role of ‗natural origins‘ in medicines
production. Plant made pharmaceuticals are distinguished from the herbal tradition on the
basis of the intervention of researchers, which establishes it as continuous with
conventional biopharmaceutical manufacture. This ‗placing‘ of biopharming has
consequences for the way in which groups subsequently come to assess the potential of the
technology. As are some researchers, some groups are less than positive about herbal
remedies, distinguishing them entirely from conventional pharmaceuticals in a way that
echoes that of Professor L.

However, for other groups herbal and botanical medicines are used to illustrate a critique of
modern society. The valorisation of herbal medicines represents a rejection of the
mainstream pharmaceutical paradigm (Calnan, Montaner, and Horne 2005). It is perhaps
unsurprising then that it is in those groups in which rejection of biopharming and
pharmaceutical biomedicine is at its strongest that herbal medicines are most valued, as in
the extract from group C below:

Catherine: When I was in the Caribbean, in Barbados, we had stuff called aloe vera,
and it was the most amazing…I got sunburn, and I used this aloe vera and it was
absolute…and this is all natural
Violet: And we don‘t take any notice of them, if you passed them and didn‘t know they
were marvels like that…
Catherine: And we‘re destroying as well so much of the Amazon rainforest, we don‘t
know, what, what wonderful plants and stuff are out there, natural, which could help
us do we?
(Group C, meeting 1)

For Catherine, the pursuit of novel pharmaceuticals in the form of biopharming occurs at
the expense of ignoring the medical potential of existing, ‗natural‘ products. Such framing
of herbal medicines represents a parallel to that of organic foods, and an alternative to the
narrative of biopharming described by researchers.

The relationship between biopharming and herbal medicines is complex, as rejection of a


narrative which links biopharming to herbal medicines can be for different reasons. For
some groups, notably A (young urban male) and F (young rural mixed), two of the younger

163
groups, herbals are unregulated and an unsuitable progenitor of biopharming as
biopharmaceutical production. In contrast, for others, particularly some participants in
Group C (older urban mixed), the value attached to herbal medicines contrasts with
negative perceptions of biopharming. Each of these is keen to place biopharming within
conventional rather than botanical pharmaceuticals, but for differing reasons.

As do expert accounts, publics struggle to reconcile the mainstream pharmaceutical features


of the technology with traditional plant-based remedies. This is related to the valence
attached to herbal medicines by group members. Those who are strongly attached to herbal
medicines are often more sceptical about the pharmaceutical paradigm of modern medicine
and are more likely to view biopharming with scepticism and to see it as unnatural.
Conversely, those who see little problem with existing pharmaceutical practice and are
sceptical about herbal medicines are likely to see no problem with the ‗unnaturalness‘ of
biopharming.

The discussions exemplified by the extracts here demonstrate the existence of a number of
foundational narratives of molecular farming that place it in relation to existing trajectories
of medical research and pharmaceutical production. These are not exclusive and discussion
in any one group often moves between them. A first narrative links molecular farming to
existing pharmaceutical practices, both chemical, and in one group, biologic. This serves to
fix molecular farming as an incremental and continuous development in drug production.
This introduces questions of motivation and focus related to industrial control of the
pharmaceutical industry. The second association builds on this, placing molecular farming
in a genealogy of medical research which includes vaccination and the discovery of
penicillin. This serendipitous narrative is particularly important in generating support for
research. Finally, group participants consider recombinant plant made pharmaceuticals as a
progression of the traditional use of plants for medicines. While little continuity is
perceived, the consequences of this for biopharming are determined more by the diverging
values attributed to herbal medicine.

Expert narratives of biopharming place it as emerging from both agricultural and


pharmaceutical research traditions. These involve a focus on either the platform or the

164
products of biopharming respectively, and exist alongside one another. For publics
however, the relevant pasts for biopharming are entirely medical and concentrate on the
pharmaceutical product rather than the agricultural platform. For both experts and publics,
pharmaceutical pasts act to authorise the promise of biopharming. Examples of past
medical research success such as penicillin are evoked by focus group participants to
highlight the serendipitous, unpredictable nature of medical progress. Experts on the other
hand evoke the success of aspirin and vinblastine to normalise plant made pharmaceutical
production in terms of botanical medicines, but also to assert their efficacy through a
contrast with herbal remedies. These pharmaceutical beginnings provide the basis for
expert and lay expectations of biopharming‘s futures.

Part II Placed Expectations

The pasts of biopharming establish the basis for the choice of technological platform and
for the association of the new technology with the ‗authority of a parallel venture‘ (Said
1975). For both publics and experts, these pasts ‗place‘ biopharming temporally and
authorise particular visions of biopharming‘s future. Biomedical technologies, and
particularly biotechnologies, are frequently developed in a ‗regime of hope‘ (Moreira and
Palladino 2005). It is on the basis of this hope and/or hype that they garner the economic
and social support through which the potential may be realised. Futures are thus
performative in the constitution of technologies in the present. However, they are not only
temporal but spatial, as they are located within imagined geographies of biopharming‘s
future. In this section I explore how both experts and publics place the future promises of
biopharming.

4.3 Experts: Philosophy and Profit?

The promise of biopharming is attached to imaginaries of existing agricultural and


pharmaceutical production. Biopharming researchers advocate its adoption as low-tech,
cheap and scalable. Its pharmaceutical promise draws explicitly on imaginaries of

165
agricultural space, as described in Ma et al.‘s (2003) aspirational introduction to the
technology:

―Imagine a world in which any protein, either naturally occurring or designed by


man, could be produced safely, inexpensively and in almost unlimited quantities
using only simple nutrients, water and sunlight. This could one day become reality as
we learn to harness the power of plants for the production of recombinant proteins on
an agricultural scale. Molecular farming in plants has already proven to be a
successful way of producing a range of technical proteins. The first plant-derived
recombinant pharmaceutical proteins are now approaching commercial approval,
and many more are expected to follow.‖
(Ma et al, 2003: 794)

This introduction lays out the products, the process and the advantages of the technology. It
presents the future of molecular farming as proximal to the research it intends to go on to
describe and places both agricultural and medical aspects of biopharming to the fore.
Molecular farming in plants is presented as a pharmaceutical development moving to
appropriate the existing advantages of agricultural production. Its future promise is centred
on its safety, its cheapness and its scale, all derived from ‗harnessing the power of plants‘.
The realisation of this promise will ―make protein-based pharmaceuticals available to
everyone who needs them, at a cost that everyone can afford‖ (Ma et al., 2003: 803).

Biopharming researchers here develop a powerful vision of a new type of global, low-
technology medicine achieved through biotechnology. This involves the performance of not
only imagined futures for biopharming, but imagined geographies of both the developing
world and the European pharmaceutical industry.

4.3.1 Imagined Geographies and Technical Solutions

The hopeful biomedical characteristics of biopharming constitute the key part of the
potential of the Pharma-Planta Consortium, as expressed in their launch press release by the
consortium‘s coordinator Professor Rainer Fischer of Fraunhofer IME, Aachen and the
scientific coordinator, Professor Julian Ma of St. George‘s Hospital, London:

166
―Infectious diseases are the leading cause of death in children and the second highest
cause in adults, says Ma. 'These diseases primarily affect people in developing
countries who do not have access to and cannot afford the medicines and vaccines
that are on sale in developed countries'. Fischer adds: 'There is a desperate need to
find ways to produce modern medicines in sufficient quantities and at a cost that
would make them available to everyone. We believe that using plants to make
pharmaceuticals could make a significant contribution.‖
(PPC 2004)

Pharma-Planta‘s product goal is the entry of an HIV vaginal microbicide for developing
world application into first stage clinical trials in the EU. This goal both determines the
shape of the technology and positions it in relation to conventional pharmaceutical and
agricultural biotechnologies. As well as representing a future vision of the technology
revolving around medicines and vaccines, there are a series of imagined geographies (Said
1995) to biopharming. The aims of the PPC are placed, not only in terms of the narratives
described in section 4.2, but through imaginaries of global disease and pharmaceutical
distribution, of the economic geographies of pharmaceuticals, of agriculture, as Ma
describes elsewhere:

"The concept of a plant-based pharmaceutical platform would bring agriculture to


medicine in a way that would allow predominantly agricultural economies to improve
their healthcare and other needs"
(Ma, Barros et al., 2005: 584)

This description of the technology places biopharming in agricultural economies. Here the
dominant promise of biopharming is ―the provision of inexpensive drugs on a large scale to
those who are currently least able to afford them‖ (Ramessar, Capell and Christou 2008:
123). Diseases and technologies in PPC imaginaries of biopharming are located - the
products of the consortium are intended specifically for developing world diseases.
Moreover, the selection of maize dictates that these agricultural economies require the
ability to process the production crop. As the same author expands in a separate review:

―plants can be grown inexpensively on an agricultural scale and, most importantly,


could allow the regional development of pharmaceutical production to match local
demand‖
(Ramessar, Sabalza, et al. 2008:410)
167
In the futures described by Ramessar, Sabalza et al. (2008: 409) and by Ma and colleagues,
pharmaceutical production and consumption are co-located, and the technical problems of
drug transport and delivery are bypassed. The placed promise of biopharming is elaborated
in an interview with Professor B. Not only does this promise orientate the consortium‘s
own activities, but distinguishes it from conventional pharmaceutical production and its
organising logic or ‗philosophy‘:

One of the Pharma-Planta antibodies has been produced in mammalian cell culture
system. You would need more than the total world's fermentation capacity for animal
cell culture just for one drug to inoculate Africa only. So pharmaceutical companies
don't want to know because Third World pharmaceuticals isn't a good profit
maker, so you're going to have to work very hard for the big guys to come on board
when you want to produce a drug at a 10th the price of the drugs that are sold in the
Western world and sell very cheaply as well. So you've got to produce cheap and sell
cheap. If you could produce cheap and sell at the same price that they sell here they'd
love it. But that isn't the philosophy behind Pharma-Planta.
(Professor B, my emphasis.)

While the technology of biopharming is continuous with mammalian cell culture systems,
the vision of the PPC separates both it and its products from conventional pharmaceutical
production. For this researcher, PPC biopharming‘s concern with expanding the availability
of biopharmaceuticals beyond the developed world distinguishes it from these other
systems.

Significantly, the PPC attempts to make this commitment to the developing world future of
biopharming material. The ‗Pharma-Planta philosophy‘ described by Professor B. is
embedded in the products of the consortium through the use of intellectual property
mechanisms. Consortium members are signatories to a Statement of Intent on Humanitarian
Use (SIHU), which describes how:

―We appreciate that the Program will lead to the generation of Knowledge [sic] that
will be important for the development of products to address health needs of the poor
in developing countries. This Knowledge may include information, copyrights,
patents, designs, plant varieties, supplementary protection certificates and other
forms of protection.

168
Therefore, we, the Consortium Members, agree that to the best of our ability, we will
make Knowledge that may be created by our Program, freely available for the
achievement of Humanitarian Purposes.‖
(PPC 2005)

The use of intellectual property mechanisms to reinforce the PPC‘s goals represents a
recognition both of the potential commercial importance of biopharming and the
consortium‘s ability to leverage access to novel technologies. However, at the same time it
places biopharming within a conventional mode of ‗technology transfer‘ rather than
capacity building in developing world healthcare. While the technology may be of
application to the developing world, research is still situated within European research and
patenting domains.

The future of biopharming as it is envisaged within PPC research is explicitly located


beyond the EU. This is developed in a 2005 review by PPC Project Manager Richard
Twyman and colleagues:

―The developed world is accustomed to readily available injections, pills and liquid
preparations, and most of us take for granted the ability to store and distribute such
formulations in the appropriate way to maintain their viability (e.g., chilled or
frozen). In the developing world, without reliable storage or distribution networks,
getting medicines to those most in need can be difficult. However, plants provide the
ideal solution to this problem‖
(Twyman et al. 2005:2)

As it is narrated by researchers, biopharming represents ‗appropriate technology‘, based on


an imaginary of pharmaceutical networks of the developing world which contrasts with the
‗taken for granted‘ nature of Western biomedicine. Biopharming thus bridges, but also
reinforces, the difference between the developed and developing world medicines. In doing
so, it outlines and attributes characteristics of medicine delivery which frame the challenges
of developing world healthcare in technical terms.

For PPC researchers, the promising social vision of biopharming relies on the establishment
of a parallel pharmaceutical innovation system based on alternative technologies drawing
on agricultural space and infrastructure. The Pharma-Planta philosophy establishes a

169
boundary between it and existing biopharmaceutical manufacture based on their technical
vision: the goal of ‗inexpensive medicines available to all‘ (Twyman et al. 2005). In
providing this expected path of development, the PPC orientate regulatory and research
efforts in a particular direction, with the explicit aim of increasing the reach of
pharmaceuticals.

Not only does the PPC vision involve the mobilisation of an imagined future, but also of an
imagined geography (Said 1995; Martin, Brown and Kraft, 2008) of relations between
European lab science, Western cell cultures and fields of future pharmaceuticals in the
developing world. The ‗translation‘ involved in the movement of a technology from
research to application, invokes an ‗imagined geography‘ of movement from the bench to
bedside (Martin, Brown and Kraft 2008; Wainwright et al. 2006), or in this case from the
laboratory to the field. This involves not only a depiction of the global context for
biopharmaceutical production, but also of the role of agriculture in developing countries.

Through a focus on the developing world, the promise of the PPC is ‗placed‘ not only in
relation to pharmaceutical and agricultural production, but also in a more narrowly
geographic sense in terms of its potential sites of application. It relies on an understanding
of pharmaceutical and intellectual property systems in the developing world which
distinguishes them from those of the West. The consortium envisage a technology strongly
linked to a particular social and geographical context of application, in which biopharming
represents a novel solution to the technical problems of pharmaceutical delivery in the
developing world. For researchers, the PPC‘s focus on this vision distinguishes them from
the mainstream pharmaceutical industry. This distance is reinforced by the selection of
targets designated as developing world diseases, and embodied in the research process
through the use of intellectual property protection.

However, the development of molecular farming is also reliant on ensuring that the distance
between it and existing biopharmaceuticals is not too great, and that continuities as well as
discontinuities can be maintained. A second vision for biopharming re-associates the PPC
with conventional pharmaceutical manufacturing. The salvational promise of biopharming

170
must be supported by equating the ‗promissory capital‘ (Martin, Brown and Turner 2008)
of existing biopharmaceutical markets.

4.3.2 Plant Made Profits?

The developing world vision of biopharming is accompanied by the expediencies of its


location with the EU and the goals of the funders of research. The intersection of economic
and other goals is at the heart of the PPC, as described in the following extract from an
interview with a member of the consortium. He introduces the ‗dual role‘ of the PPC as a
negotiation between EU and individual visions:

The PPC was basically funded by the EU under its 6th RTD programme, so that‘s
you know research and technology development. Now the reason the EU funds these
programmes is to improve the competitiveness of Europe on the global scene. So the
bottom line is, they're funding this research to translate this, so that this can be
translated into money for small biotech firms within Europe. So they're funding the
PPC, the PPC is mostly academic universities, but I guess part of the reasoning of
the EU is, if they fund such a consortium, which also includes a few small biotech
partners, this will ultimately translate into profits, you know, for the pharma
industry through agricultural biotechnology.
(Dr E., my emphasis)

The first introduced goal of biopharming research is as a generic biopharmaceutical


production platform, tied to an EU agenda which links technological and economic
development. For the Framework Programmes within which the PPC is funded, an
important goal is to enhance the contribution of European research to economic growth
(EU 2002). Biopharming‘s promise in these terms relates to the economic significance of
conventional pharmaceutical production. Underpinning this approach is a second version of
technological transfer - not from developed to developing world, but from ‗bench to
bedside‘ in a linear translation from academic research to innovation and novel products.

The promise of biopharming is associated with its potential contribution to the European
pharmaceutical industry. The development of biopharming represents an opportunity to
overcome the shortage of supply from conventional production in the face of rising

171
demand. The economic potential of this is immense, as described in the Biotechnology
Industry Organization‘s introduction to PMPs

―In January 2005, market research firm Frost & Sullivan predicted that the plant-
made pharmaceutical market could realize revenues of $98.2 billion by 2011‖
(BIO 2006)

Frost and Sullivan‘s estimate now seems optimistic. While powerful in mobilising public
and private investment, the $98.2 billion market looks unlikely to be realised by 2011. The
second vision of biopharming presents it as discontinuous with existing
biopharmaceuticals, as a technology with greater reach, as a low technology, low skill
production platform. The researcher continues by describing how the approach of the
consortium is shaped by this second imperative:

I mean in the initial grant proposal which went to the EU and the feedback from the
EU, there was an element of important molecules for global health. So it’s a dual
thing. One is to build these basic platform technologies within which you can develop
systems for any molecule, not necessarily just for developing countries, not
necessarily just for TB, malaria, and tuberculosis. But in its research and
development phase, what PPC is doing, and I think this is where critical thinkers like
Julian Ma and Rainer Fischer were influential, they decided that the best molecules
to apply initially for these platform technologies were these emergency molecules.
Tuberculosis, malaria, AIDS, lets develop molecules for these diseases. OK?
(Dr E, my emphasis.)

The success of the PPC‘s ‗critical thinkers‘ is in achieving a rapprochement between what
Sunder Rajan (2006) describes as the competing tropes of ‗nation‘ and national
competitiveness (to the extent that this can be used for the EU in this context) and
‗salvation‘. Social and ethical considerations have been engineered into the technology
itself and form a crucial part of its imagination (cf. Franklin 2001). The generic platform
approach of the EU is co-opted by a researcher vision of the importance of particular
avenues of research. The consortium is attempting to combine the economic future of the
EU with the development of medicines for the developing world. In concluding the
description introduced above Dr E., an expert in intellectual property, describes how this

172
approach will lead to a future of biopharming benefiting from a synergistic coupling of
developed and developing world applications:

So the molecules probably will be developed at the end of this first phase of PPC, but
as far as the main role of PPC in the EU is concerned, at the same time as
developing these molecules they would have developed these platform production
technologies which would be applicable to any molecule, whether it‘s for the
treatment of atherosclerosis, diabetes, whatever, you know, diseases that affect both
the West and the East
(Dr E., my emphasis)

The PPC is underpinned by a hopeful vision of the future in which financial and social
concerns come together around a single technological platform. The success of the PPC in
the developing world, in terms of specific products, is accompanied by global success
based on the agricultural platform. The spatial and technological imaginaries of
biopharming are reconciled by the end of the project. In this Pharma-Planta member‘s
description, the coupling of products aimed at developing world diseases, the ‗emergency
molecules‘ described, with economic development, is an integral part of uniting the
commercial and humanitarian motives of biopharming. PPC researchers position
biopharming as a technology of both economic and social potential of benefit to both
developed and developing worlds. However, it remains difficult to square the ‗Pharma-
Planta Philosophy‘ with the promissory futures of biopharming for EU pharmaceutical
companies, and with longer term aims to develop biopharming as a platform technology.

While the ‗Pharma-Planta Philosophy‘ locates the promise of biopharming outside the EU,
it is brought back in by its funding and institutional locations. To over-simplify, the
‗symbolic capital‘ and social value of biopharming is located apart from its economic
value. Researchers do not resolve this potential conflict, but instead attempt to co-opt
European pharmaceutical goals in pursuit of developing world medicine.

4.4 Publics: Pharmaceutical Promises

Hopeful visions for agricultural biotechnology, including the introduction of Golden Rice,
were met with scepticism by European and British publics. In contrast, public discussion of
173
biopharming in focus groups is far more accepting of the technology‘s promise, and
suggests the emergence of a widespread ‗community of promise‘. However, discussing
biopharming in relation to existing interventions of medical and agricultural technology
‗places‘ this promise in different terms to those adopted by researchers.

4.4.1 Technical Promise and Social Failure?

The goal of the PPC is that of producing a cheap and affordable pharmaceutical for
application in the developing world. This is reflected in the examples of biopharming
provided to focus group participants, and a vision of improved medical care underpins the
support for molecular farming that they express. Participants were presented with a booklet
describing molecular farming and detailing three possible applications, the two initial PPC
projects, HIV antibodies in maize and rabies antibodies in tobacco, along with gastric
lipase, also produced in maize.

All three examples given to groups were received positively, but each was discussed in
varying depth and detail. As Sam from Group F (young rural mixed) put it:

Sam: None of them were doing a bad thing were they, or potentially doing a bad
thing, I think the HIV would potentially be a massive thing in certain areas of the
world wouldn't it, if you could do something about that. All good I thought.
(Group F, meeting 2)

The formation of imagined communities relies on shared information and representations


(Anderson 1983). It might therefore be expected that in the absence of such shared
information, the formation of such communities would be hampered. Such appears to be
the case in focus group discussion of biopharming. Of the three examples, UK broadsheet
newspapers carried 2,300 stories of all types involving HIV between 2006 and 2008, while
just 188 stories involving rabies and 298 involving cystic fibrosis are found in a search of
the same media over the same period19. HIV is prominent in Western media discourse, and
all groups were able to discuss the scale of the crisis. In contrast, almost no discussion of

19
Based on search of Nexis UK database of UK newspapers, November 2008
174
the cystic fibrosis example takes place, while the discussion of rabies in Group D (older
rural female) highlights the problems of a lack of information:

Molly: well I‘m surprised how many people are killed by rabies, 50,000, it’s not
really what we come across in this country is it? If you could get a vaccine for that,
antibodies…
Sofia: rabies, is it just from animals?
RM: yeah, dog bites and things
Elsie: Is it just dogs, rabies?
RM: and bats, foxes things like that
Molly: but 50,000 is a lot of people, I don‘t know how many die from HIV but, I don‘t
suppose you can count that, because it‘s terrible for the children who inherit it
Elsie: do people get it in England, rabies?
RM: I think there was one person last yea
(Group D, meeting 2)

There is confusion among group participants about the promise of plant derived rabies
antibodies. Information has to be provided by the facilitator about both the aetiology and
epidemiology of rabies. Groups generally discussed this example only briefly, and contrast
the number of deaths described compared to HIV, even when considering rabies as a
serious problem.

The HIV microbicide prompted by far the greatest response in group discussion. Indeed, in
some groups it framed the entire conversation around biopharming. Discussion in the
groups is often initially strongly positive as for Evie in Group B (young, urban female):

RM: What did you think the most interesting thing was that you read?
Evie: It could treat AIDS, it could be used to treat AIDS, it‘s a very big problem and
a very big issue, so it‘s not about, flu, I mean flu is a big issue as well, but not
something that has an effect on so many people
(Group B, meeting 2)

The concentration of PPC research around the core project of HIV antibodies in maize is
paralleled by the positioning of HIV as the core hope for biopharming in focus group
discussion. The support evinced for the production of HIV antibodies draws on similar
notions of progress through biomedicine as for experts. The promise of the technology is

175
also clearly related here to the scale of the problem. In Group E (older rural mixed), a
similar discussion is led by Jane:

Jane: You look at rabies, and I mean HIV, it‘s an absolute plague on humanity isn‘t
it, and if you can, I would, basically, I would like to see something working along
these lines, but I think it‘s a long way down the line and it‘s got to have a lot of
controls, and there‘s somebody got to be a great PR guru somewhere has got to
really sell this in the right way so that it doesn‘t meet with what happened with the
GM crop trials, and you know, scaremongering with your headlines in the press and
all that sort of thing
Joan: I think you‘ve also got to start with more education at the beginning.
Jane: I mean it‘s a long term process
Joan: They don‘t want to stop, certain areas don‘t want to stop HIV do they?
Helen: Well it makes you wonder
Douglas: It‘s a good point actually, probably more deaths occur through political
slaughters than there are through people dying of disease, so in the scheme of things
you know, where does it lie? I don‘t know
(Group E, meeting 2)

The salvational promise of biopharming is introduced by Jane in terms of the sheer scale of
HIV infection, which creates an imperative to move biopharming beyond the compromised
potential of GM. However, for other participants there is overt scepticism about the ‗magic
bullet‘ potential of the new technology, linked not so much to imagined geographies of
pharmaceutical distribution, but more to imagined geo-politics. Unlike in the discussion of
researchers described above, the primary problems of developing world healthcare are not
seen as technical, but as social and political. Both the promise and problems of
biopharming are consequently not those of the technology itself, but of the context in which
it is placed.

Both expert and lay accounts attribute characteristics to the countries, populations and
governments of the developing world in articulating their understandings of the promise of
biopharming. For publics, the social and political context into which the applications of
biopharming enter is key to assessing, tempering and ultimately deconstructing their
promise. The understandings of biopharming‘s future are further illustrated in the following

176
discussion in Group D (older rural women) in which they move to position the potential of
the HIV example:

Sofia: And there again, a cream, would they use it? I mean, I mean, there‘s such a
thing as using something to stop you getting it
Molly: Of course there is
Sofia: And it‘s far cheaper, but they don‘t use it you see, so I don‘t think that‘s really,
I don‘t think, in the African countries I‘m meaning, they don‘t,
Vera: Well they‘re not educated enough
Sofia: They‘re not educated enough
Molly: They don‘t want change
Sofia: No they don‘t
(Group D, meeting 2)

Expectations of the PPC microbicides are related to a cynicism derived from existing
attempts to restrict the spread of AIDS through the use of condoms, rather than a promising
pharmaceutical imaginary. Their technical potential is tempered by continuity with the
failures of past efforts and the imagined characteristics of developing world populations.
The promise of biopharming to offer cheaper, more widely accessible medicines is
challenged on the basis of the perceived characteristics of African populations.

However, the tempering of the promise of biopharming relates not only to the social and
political context of its application, but also to the technology itself. The scepticism
described here parallels the findings of the AEBC‘s non-food agriculture study, which
described ‗a huge mismatch‘ between public expectations and what the HIV case study
appeared to be offering (CWR&D 2005). This can be seen in Group B, in which the initial
enthusiasm of Evie is tempered through a deconstruction of the pharmaceutical approach:

Jo: I mean when you read 40 million people, that kind of makes me think its such a
huge, huge, huge problem that it kind of makes me think it‘s quite desperate really
isn‘t it and kind of do whatever you can,
Sarah: just making me think back to what we were talking about last week, you know,
about how doctors and things are just handing out antidepressants sort of, for
anybody, who, or antibiotics, is this going to be a, you‘ve got HIV, apply this cream
it‘ll be alright, or is it not really addressing the issue
(Group B, meeting 2)

177
In this extract, Jo introduces a moral imperative for some form of response to HIV, not as
support for the technology itself, but for its goal. Again, it is a case of trying anything,
rather than necessarily being motivated by the promise of biopharming. Sarah‘s more
sceptical responses highlight that this may not necessarily be the best response, and may in
fact be ineffective. Here she starts to question the promise of medicines in general. Rita
elaborates this as group discussion develops:

Rita: I suppose the approach of curing disease will always be a necessity, but does
this sort of put prevention on the back burner? I suppose that‘s not totally what we
are considering here, but er, there is, there has been, over the, you know, and even
more so in modern times, the need to focus on cure rather than prevention, because
of our lifestyle, disease and illness seem to be on the increase, and it would be, I think
it would be very heartening if somehow we could increase the emphasis on
prevention somehow
(Group B, meeting 2)

Rita introduces a challenge to the approach to medicine represented by biopharming


situated in a concern about the overall focus of pharmaceutical research. Her response
situates biopharming within the group‘s first discussion, which challenged the efficacy and
desirability of the conventional/industrial pharmaceutical model. As well as a challenge to
biopharming, this is part of a wider challenge to the promise of this pharmaceutical
paradigm. Unlike for researchers however, biopharming sits within existing pharmaceutical
production, and as it does not offer a cure, the technical promise of biopharming is not
sufficiently ‗forceful‘ (van Lente 2000).

A more strident rejection of the pharmaceutical imaginary underlying biopharming


developed along similar lines is expressed by Catherine in Group C as she discusses rabies:

Catherine: Wouldn‘t it be better to treat the dog rather than…to treat the cause
rather than the symptoms, wouldn‘t it be better to get rid of it. Because we don‘t have
rabies in this country do we? We‘ve eradicated it; we don‘t allow pets in without
quarantine, do we? So we don‘t actually have rabies in this country. Wouldn‘t it be
better to channel our energies to dogs or animals that are carrying rabies rather
than…
RM: So it‘s a question of focus?
Catherine: Yeah, I think we‘ve gone completely wrong…
178
Peter: In reality, it could well be by bats or birds I mean, there‘s always possibilities,
the countries which have it, it‘s not just dogs…
Catherine: No, there‘s bats as well, I know I know, but I‘m just trying to say is that
we‘re treating the symptom rather than the cause and this is sometimes where we go
wrong. It‘s like HIV, unless you treat, unless people you know do something about
their behaviour, then there wouldn‘t be all these problems you know, this is, all I‘m
trying to say is let‘s tackle the cause rather than the results of what happens, it‘s
always cause and effect isn‘t it?
(Group C, meeting 2)

As in the discussion from Group B, for Catherine, the problem with the products presented
to the groups is not in their potential efficacy, based on assumptions about the spaces of
application or of the failures of past efforts, but is in their association with an unwanted
model of biomedicine. Elsewhere in group discussion she suggests the association of
medicine with science has led to ‗unrealistic expectations‘ arguing that

―we‘re just making people expect far too much, and it‘s all the fault, I believe, of
scientists and everything. I mean ever since they‘ve cracked the human gene, people
expect them to cure [everything]‖
(Group C, meeting 1)

Although, no other group rejected the ‗biomedical imaginary‘ (Good, 2001) so strongly,
placing molecular farming within a narrative of a misguided society echoes a number of
groups‘ discussions around first generation genetically modified crops. Marris et al. (2001)
note in their research on first generation GM that ―when GMOs were singled out [by
participants] for special attention, it was because they were felt to crystallise a particular
view of the world and way of life that they were not entirely happy about‖ (2001:83).
Genetic modification represents a significant step involving "unnatural" modification and
transformation, as opposed to ‗working with‘ nature. They highlight how this perception of
agricultural biotechnology is grounded in an understanding of existing agricultural practice
as unnatural, and, in the UK in particular, of support for more ‗natural‘ organic production.
Similarly, the comments of Catherine and Rita suggest that narratives of biomedical
innovation are not universally positive, and reintroduce the relation between biopharming
and the alternative narratives of botanical medicines.

179
In considering biopharming, group discussion replaces an imaginary of global
pharmaceutical distribution with one of local socio-political health contexts, and challenges
the focus of modern medicine on treatments rather than cures and prevention. Finally,
Group F (young rural mixed) considers how biopharming may be transferred from
European research to its application in the developing world:

Rebecca: Hmmm, no, I was just wondering, I don't think so, I was just wondering
whether, if it‘s still kind of the Western thing that‘s still developing everything and
the effect would it still get through properly, or this, has it made it sound like, oh you
know for AIDS, with the huge numbers in Africa that might benefit from it, would they
benefit from it, or would it be kept for us
Max: I suppose they've got a much better chance of it haven't they
Rebecca: Yeah I guess so, because it‘s cheaper
Max: And eventually it will filter through to them, but yeah, I'm sure we'll benefit
from it first
(Group F, meeting 2)

The limits on promise are here not a result of the context of application, but of its
production, of the logistics of transferring knowledge and technology, and the belief that
Western technology would benefit Westerners first. This extract displays an understanding
of scientific research as strongly localised and imperfectly relayed, complicating the
narration of biopharming by researchers that relies on a relatively uncomplicated linear
model of translation from European research to developing world application. Rather than a
direct translation, here participants subscribe to a ‗trickle-down‘ model, despite the overt
focus on a disease which, as described in both lay and expert narratives, is seen as a
predominantly developing world issue.

The ‗salvational‘ public health promise of biopharming is important in enrolling public


support, but suggest that this support is conditional upon placing the technology within a
broader socio-political context. For groups, the problems of developing world healthcare
are seen as social as much as technical, and there is concern that biopharming will become
another failed technical solution. This assessment is strongly dependent on the presence of
imagined geographies of the developing world and their socio-political status.

180
4.4.2 Profits and the Purpose of Pharmaceutical Production

Initial assessments of the potential of biopharming described by focus group participants


are overwhelmingly supportive. This relates to a salvational narrative of biopharming in
relation to developing world disease and a sense of moral imperative and responsibility.
However, as for researchers, other factors complicate this narrative, not least a discussion
of the economic promise of novel pharmaceutical technologies. In continuing Group B‘s
(young urban female) discussion of biopharming, Sarah introduces the potential costs of
pharmaceutical development as a potential restraint on production:

Sarah: Well I‘m just thinking about how much these things cost to trial to make, to
research, all this sort of thing, and it‘s saying here that that this cream will be ready
for 2009, well, how many people will be affected by then … it‘s more of a hope, we‘ll
hope that it works, we hope that the clinical trials will go through, but it might not, I
mean how many things go through clinical trials and then are never seen again? Like
Rita was saying, it‘s, where all this is going? I hope, I mean I don‘t know what is
happening, but I‘m hoping while all this is going on, somebody is thinking about a
preventative measure as well for if this doesn‘t help, or doesn‘t go through, which
I‘m sure they are, I‘m sure this isn‘t the only thing…
(Group B, meeting 2)

The tempering of promise is not only a judgement on the technology itself, but also on the
people and societies within which it is produced. As seen above, the promise of
biopharming is situated by participants in the context of imagined characteristics of the
places and people for whom it is proposed. However, these spaces of prospect exist in
relation with those of innovation – the challenge is one not only of application, but of
translation and indeed of the research process itself. Sarah‘s understanding of the hope of
biopharming is grounded in relation to her understandings of medical research in general
and the rate at which new pharmaceuticals fail clinical trials. While not opposed to the
development of biopharming, she is sceptical about its promise. Unlike earlier discussions
which situated the future challenges of biopharming in relation to imagined characteristics
of the developing world, these limitations are those of developed world science and
industry.

181
Enrolment in a ‗community of promise‘ involves credence in other actors and trust in their
authenticity of purpose (Brown 2007). Where such credibility is present, support for
biopharming is strong, including for economic purposes:

Jane: Well research on this scale obviously costs a lot of money, and big institutions
are not going to put their back into it if they…this is why they license the products so
they get some payback isn‘t it, before it then goes…and you can use it as a generic
tablet or whatever
(Group E, meeting 2)

The economic promise of biopharming is seen by Jane as a just reward for the pursuit of
social goals. Economic and social goals are here entwined and their realisation linked.
Unlike the challenging of the pharmaceutical paradigm described above, for Jane the
pharmaceutical innovation system is effective. However, her views are not shared by all in
the group, as the economic promise of biopharming contributes to establishing or
challenging the credibility of information about the technology. As described in Chapter
Three, groups were presented with a series of statements related to biopharming. When
presented with a quotation from a PPC paper describing the potential of the technology20,
Group E (older rural mixed) question it on the basis of ‗hidden agendas‘ based on profit:

Patricia: No, so if somebody‘s out to make money by convincing you that something
will work, I would question it
Joan: I think I would question everything
Patricia: Well, some things more than others
Jane: Yeah, but you‘re looking for hidden agendas here aren‘t you
Patricia: That‘s right
(Group E; meeting 2)

The implication of this discussion is that the believability of claims about the potential of
biopharming is linked to scepticism of commercial motives. Despite Jane‘s description of
the ‗just deserts‘ of pharmaceutical innovation, the perceived economic promise of
biopharming nevertheless impacts on the credibility of statements about it. This scepticism
represents a sociologically aware approach to hype and expectations around new
20
The quotation from Ma, Barros et al. (2005) states that ―250 acres of greenhouse space would be sufficient
to grow enough transgenic potato plants to meet South East Asia‘s annual demand for the hepatitis B virus
vaccine―
182
technologies. The linking of credibility to profit, or lack of it, is elaborated in Group A‘s
discussion of the same quote:

Mark: We‘re more likely to believe it because it‘s a public sector organisation, even
though they‘ve clearly got an interest in making claims about their research, because
of their reputation and prestige and those sort of things,
Ed: There‘s not a profit motive is there, which I think occasionally…
(Group A, meeting 2)

While the discussion of the potential of the PPC envisages a synergy between European
wealth and developing world health, for focus groups they are potentially conflicting – not
necessarily in terms of the technology itself, but in terms of the authenticity of possible
futures. For Group A technological promises made by public sector organizations such as
the Pharma-Planta Consortium are seen as inherently more believable. For biopharming,
the mobilisation of a community of promise in Europe is enabled by representations of
researcher motives, as well as perceived social benefits.

The link between particular promises and the willingness of publics to support technologies
is highlighted by a highly critical discussion in Group C:

Patience: I think that my main concern is feeding us plants with medicines in. Are we
sure what we are putting inside us? You know, the long term effects and that? If we
are not sure, then why are we doing it in a vast scale, are we doing it for money or
for humankind? That‘s my concern, why are they, the pharmaceutical companies and
the agricultural people, doing this in a mass scale, is it for money or is it for people
to get better?
Violet: I think it‘s for fame and for money as well, because once they get going, it‘s
just like an addiction, they want to prove your ingenuity in everything, so you try to
help, but you are clutching onto your fame. Nowadays, everybody, or anybody at all,
can just get chalk [calabash chalk], and put it in a bottle and say that, oh, it‘ll do
something and it‘s wonderful…people are producing drugs without being checked
and things like that. I put it down to money and selfishness. In the news they went and
found people with machines producing paracetamol or things like that, it‘s just for
money, and sheer wickedness, they don‘t care, it‘s greed.
(Group C, meeting 2)

183
Patience‘s initial worry is for the motives underlying the development of plant made
pharmaceuticals. She is concerned that the economic promise of the technology will drive
its development too rapidly. However, Violet links this to broader concerns about the
motives of pharmaceutical production, in a discussion which echoes some of the negative
commentary of experts. Drawing on the example of calabash chalk21, she emphasises the
profitability of pharmaceuticals and the potential for subversion of quality regimes, of
checking and testing. In doing so, she introduces the importance of placing biopharming
within past narratives of promise and failure.

Distrust and Agricultural Biotechnology

The discussion heretofore concentrates on the role of pharmaceutical promise in group


discussion of biopharming. However, considerations of commerciality also allow groups to
place biopharming in relation to, and distinguish it from, the contested promise of
agricultural biotechnologies. Equally positive projected visions for the developing world
emerged in the promotion of genetically modified agriculture. However, group discussion
of these visions often represents a total inversion of the promise described, however
sceptically, for biopharming:

Will: Are you just talking about the research or about the whole commerciality of it,
the dependency on companies creating, because it‘s quite a big conversation when
you say GM, it needs to be broken up into parts. Some parts, in terms of sort of
genetically modified, I mean as you said, this sort of thing's been going on in a form
for hundreds of years, but now when we talk about GM, we're not just talking about
genetically modifying crops, but we're also I feel talking about controlling the
distribution of certain types of crops in certain parts of the world and making people
dependent on certain companies. I feel that's entered the argument as well
(Group A, meeting 1)

Will describes the potential impact of GM critically, suggesting that agricultural GM crops
are inextricably tied to commercial goals. Unlike for biopharming, the commerciality of
GM detracts from its social promise. GM represents a relationship of control and
21
Calabash chalk is a type of chalk used particularly in Nigerian and West African communities to alleviate
morning sickness in pregnant women. In the UK, the Food Standards Agency has warned that the chalk
contains high levels of lead, and at the time of the group meeting, warning leaflets were being distributed in
Hackney.
184
dependency between Western biotechnology and the developing world, and there is a
palpable lack of trust in both agbiotech companies and governance (cf. Jasanoff 2005).
Throughout group discussions, this is most clearly elaborated in terms of ‗Terminator
seeds‘, or genetic use restriction technologies (GURTs). As this discussion continues,
Group A (young urban male) consider the role of power and money and the creation of
dependency:

Mark: Yeah, what are they doing now, they're making rice that doesn't reproduce or
whatever, and then selling it so you're dependent on the rice company.
Will: Yes, exactly that sort of stuff, so you might find a farmer or a group of farmers
become dependant on the supplier, so there's an extra commercial aspect to it, which
today I think is inescapable in discussion about GM crops
(Group A, meeting 1)

Will‘s comment highlights how GURTs represent the perceived inseparability of GM from
economic concerns and the increasingly effective capitalization of agriculture through
biotechnology. They concentrate discussion of the spatialised potential of agricultural
biotechnology. Along with Dolly the sheep and genetic testing for disease, terminator
technologies were one of the most common examples of biotechnology raised
spontaneously in focus group discussion. Indeed, given that Dolly was named in the
recruitment flyer for groups, GURTs were the example shared among most groups,
suggesting that they represent an important shared feature of public discussion of GM and
of the imagined communities that form around it. GURTs were discussed in three of the
first group meetings, by Groups A [young urban male], C [older urban mixed] and E [older
rural mixed]. Group E relayed their knowledge about terminator technologies in the GM
debate as follows:

Jane: One thing I, oh, some time ago when there was the thing in this country about
whether Monsanto would get licenses for doing field…
Paul: trials
Jane: trials. And there were some, quite a few articles in the press, again, in the press
so you take it with a pinch of salt perhaps, one thing I picked up on was, where they
grow, say tomatoes, it might not have been, but a particular crop, with the GM seed,
the firm provide the seed because the farmers are renting the land from them they‘re
in hock to the chemical firm if you like, and because it‘s a chemically altered seed
185
that they‘re using, it‘s sterile, so whereas traditionally a farmer would grow wheat
and he would save so much to have the seed, I‘m talking about small farming
communities now, not your major combines, but in the Third world countries, this is
still a major way of life, but they haven‘t got the seed to carry over to the next year,
so they‘ve got to go back and buy more seed from this company, and that aspect of it
very much concerned me.
Paul: Yeah, that is a very valid point is that
(Group E, meeting 1)

Group E are predominantly positive about GM crops in general and biopharming. However,
terminator technologies are inextricably linked with the relationship between Western
corporations, explicitly Monsanto, and developing world farmers. Information about
terminator technologies comes from the media, and its reliability is questioned. However, at
stake is the control of farming and imagined traditional ways of life, with the risk being the
loss of independence. The issue of dependence was important to this group, and particularly
to Jane. Nonetheless, it was not simply Jane who directed this agenda, as it was picked up
by a number of other group members, and is replicated in the other group discussions
below. The link here between independence, corporate control and developing world
agriculture highlights how debate around GURTs overlaps with larger concerns. Group C
discuss terminator technologies in a similar way:

Peter: I don‘t like these GM crops where they don‘t produce seeds, and therefore the
local growers, in developing countries particularly, they‘re dependent on big food
companies to buy the next lot of seeds from, that seems to be kind of not right. But
even this I have to say, if we‘ve got serious problems with world hunger, that‘s the
only way, then I would be willing to explore it.
Violet: Yes
Peter: As with all these things, it‘s only when it‘s actually necessary
(Group C, meeting 1)

For focus group participants, GURTs act as an intermediary between ‗big food‘ and the
developing world local grower. They are not an innocent intermediary, but carry corporate
power into areas where it previously could not reach: they provide a biological means of
controlling intellectual property (Bowring 2002). GURTs place agricultural biotechnology
in a relation with the same geographies as those described for biopharming, in a

186
significantly different tone, but in the same terms of a concentration of technological power
in developed countries. Peter‘s rejection of terminator technologies and ‗dependence‘ is
qualified by the question of ‗necessity‘. By leaving the door open to GURTs in this way, he
prefigures the re-emergence of GURTs within discussion of biopharming.

In the context of biopharming, the information booklet described GURTs as a form of


containment by the use of ‗sterile plants‘. GURTs provide an instructive example of the
way the promise of biopharming is co-produced with new narratives of biotechnology.
Overall, a critical discussion of GURTs as ‗terminator technologies‘ in the first group
meeting is replaced by a promising discussion of ‗sterile seeds‘. In the extract below,
members of Group F are strikingly confident in the efficacy of biological containment and
of engineered sterility:

Sam: Wasn't it that some of these things couldn't be spread though


Rebecca: You're right, some of those self-fertilise, not self...
Max: Yeah, self-pollinate. Or sterile wasn't it?
Sam: So in that case, I wouldn't have any problems whatsoever. If you could
guarantee that, it would be tickety-boo wouldn't it. You could just go for it
(Group F, meeting 2)

In contrast to discussions of first generation GM, there is no consideration of the broader


socioeconomic context of developing sterile plants. Other groups developed the discussion
further by questioning the possibility of the ‗guarantee‘ required by Sam. While not
rejected in the same way as terminator technologies, the positive and certain characteristics
of biological containment and sterile varieties were opened to question. Returning to Group
A, conversation around biological containment methods was developed in the second
meeting. Here however, the focus was not on the economic aspect of genetic modification,
but on efficacy and controllability:

Rob: Infertile varieties, that sounds the most sensible, if I was to choose one, I'd say
that infertile varieties are the best
Euan: I'm not so...wouldn't infertile varieties require just like one mutation and
you're in trouble
(Group A, meeting 2)

187
Unlike the spontaneous discussion of terminator technologies which arose in the first group
meeting, the discussion above took place after participants were presented with a list of
proposed containment measures and asked to judge them. While the discussion above links
GURTs to corporate dominance and an imaginary of industrial agriculture, this exchange
considers them in a narrowly technical sense. Suspicion of the technology derives not from
their efficacy in extending the reach of corporate control of agriculture, but instead from
their inefficacy in restricting the spread of pharmaceuticals.

The potential of medical research is tempered by awareness of commercial pressures.


While the PPC‘s research is seen as less problematic than some, there still exists distrust in
the motives behind pharmaceutical manufacture. To place this distrust in context however, I
have compared public discussion of biopharming with that of first generation genetically
modified crops. The example of ‗Terminator technologies‘, which featured prominently in
group discussion, emphasises that concerns about biopharming differ from those around
first generation GM. The perception of an imperative for the use of GURTs to contain
pharmaceutical crops distances it from its previous role as an extension of corporate power.
McAfee (2003b) dismisses this containment reframing of GURTs as hubris. However, the
discussion here indicates that controversial agricultural technologies may have new
pharmaceutical futures discontinuous from their GM past.

4.5 Summary and Conclusions

The key findings of this chapter are summarised in table 4.1. The PPC vision of
biopharming involves the production of low cost, low technology, large scale
biopharmaceuticals for the developing world using maize as a production platform. This
vision distinguishes PPC biopharming, but co-exists with a second narrative which re-links
the technology to conventional pharmaceutical manufacturing. Neither herbal medicines
nor agricultural research feature prominently in the promise of biopharming, yet this
promise draws on the familiarity of plant derived medicines and on traditions of
agricultural research and production.

188
Figure 4.1 Summary of Chapter 4
Futures Pasts
Described in terms of both biotechnology and
Promise placed in the developing world and botany
in European pharmaceutical industry.
However, PPC ‗philosophy‘ distinguishes Tradition of herbal medicines provides
biopharming from other biopharmaceutical authorising narrative for use of plants, but
manufacture potentially problematic for perception and
regulation of pharmaceutical products.
Experts

Based on imaginaries of global


pharmaceutical distribution, developing Biopharming primarily a development in
world healthcare problems treated as biopharmaceutical production
technical question of production.
Agricultural research and experience provides
assurance of safety; research knowledge and
infrastructure; production expertise and
infrastructure. Each of these is located.

Promise placed in the developing world Relevant pasts for biopharming dominated by
through imaginaries of global health. medicine. Agriculture is absent from discussion

Anticipated medical promise limited by Authorising events in the history of medicine


anticipated social problems and difficulties emphasise the benefits and unpredictability of
Publics

of technology transfer. medical research

Pharmaceutical framing corresponds with The link between biopharming and herbal
change in attitude to GURTs medicines is challenged, but the consequences of
this differ between groups, depending on their
views of herbal medicines

Imagined geographies of health and of technology transfer underpin researcher discussions


of the promise of biopharming. The promises of PPC biopharming are placed. They relate
specifically to the application of the technology in the production of pharmaceuticals for
those outside the EU, yet the research itself and its economic promise are located firmly
among EU funding structures, regulations and publics.

PPC researchers‘ narratives of biopharming position it as a technical solution to developing


world healthcare problems, and as a new platform technology for the European
biopharmaceutical industry. In contrast, public expectations of biopharming are tempered
by a focus on the socio-political challenges of HIV treatment. They re-emphasise that
189
biopharming does not represent the first attempt at intervention, but that it is continuous
with previous efforts which have failed. Nevertheless, despite scepticism about the
particular promise of PPC biopharming, there is general support for the technology
emergent from its association with the authority of medical research in general.

Support for biopharming as medical research relies on stories of past successes, and
particularly the story of the ‗happy accident‘ of the discovery of penicillin. The prominent
role of serendipity in stories of scientific advance has been discussed by Merton and Barber
(2004). However they do not consider claims about luck and chance in scientific advance as
rhetorical devices (Sismondo 2004). In fact, the story of penicillin plays an important role
in authorising the promise of biopharming – not just as another ‗hopeful breakthrough‘
(Brown 2000), but as the basis for allowing medical research and researchers leeway in the
pursuit of unpredictable progress.

Descriptions of the operation of expectations in the constitution of stable socio-technical


assemblages of new technologies identify the development of ‗communities of promise‘ -
diverse constituencies united in the pursuit of technological visions. Imagined futures
facilitate a shared sense of community to develop among otherwise distant groups (Martin,
Brown, and Kraft 2008). This chapter has examined how the vision of biopharming laid out
by the PPC is interrogated and understood. I have described points of convergence and
divergence between public and experts in their narrations of biopharming. However,
constituting the promise of biopharming involves looking beyond rhetoric and narrative to
consider how this promise is embedded in the materials of biopharming, and how these
relate to conventional pharmaceutical products and foods.

190
CHAPTER 5: BIOPHARMING MATTERS

The enrolment of publics in communities of promise and the establishment of promising


pharmaceutical narratives of biopharming is not enough to ‗place‘ them in relation to
agriculture and pharmaceuticals. Instead, as did the products of agricultural biotechnology
described by Jasanoff (2005), the products of biopharming must also ‗make a place‘ among
existing foods and medicine. In order to do so, the relevant qualities of existing products
must themselves be established. The first task of this chapter is to explore how meanings
are attached to the materialities of foods and pharmaceuticals. I consider the perspectives of
biopharming researchers, as the ‗producers‘ of plant made pharmaceuticals, and of focus
group participants as food and pharmaceutical ‗consumers‘. The second task is to examine
how these materialities underpin or challenge the narratives described in the previous
chapter, and help to shape the promise of biopharming.

In Chapter Two, I described how the materials of technoscience both embody expectations
and are the means through which these expectations are encountered, grasped and judged.
Drawing on actor-network theory inspired work in both STS and geography, I described
how the meanings attached to these materials are contingent and are constructed through
the establishment of association and relation between actors. Bringing this work together
with the narratives of biopharming allows my discussion of the ‗placing‘ of biopharming to
be extended beyond the narrative enrolment of publics.

I first consider the role of naturalness, appearance and institutional trust in classifying the
promise of the products of agriculture and pharmaceutical production. Contrasting public
discussion of the materials of foods and medicines is important in considering how the
products of biopharming are ‗placed‘, not least through regimes of quality and safety. In
addition, the differing roles adopted by participants raise questions about the comparability
of ‗red‘ and ‗green‘ publics themselves.

191
I then return to biopharming, and examine how it produces new discourses, knowledges
and biologies of foods and pharmaceuticals, and is itself shaped by its interaction with
these. I describe how the aesthetic and material characteristics of PMPs are central to public
and experts ‗placings‘ of them and to understandings of their promise. I suggest that for
experts, combining the materials of foods and medicines contributes to establishing the
safety of biopharmaceuticals, but also challenges their efficacy and their ‗authenticity‘. For
publics, I suggest that expectations of pharmaceuticals are linked to particular product
forms and to the information and institutions that accompany them. Consequently bringing
pharmaceutical and food matters together in the case of biopharming presents not only a
challenge to food safety, but to the pharmaceutical promise of the technology.

5.1 Experts: The Promise of Safe Products

The materials and natures of foods come to embody the promise of biopharming. Their
perceived safety of foods forms an essential part of the selection of production platforms
for biopharming, and underpins the promise that separates biopharming from existing
biopharmaceutical production technologies. In particular, this promise is asserted through
the characteristics and ‗inherent safety‘ of food crops, and as in earlier debates around
genetically modified crops, much of this centres around maize.

The story of maize has been at the heart of debates around first generation genetically
modified crops. Molecular farming‘s promise of developing world medicine is also based
on the characteristics of food, in particular those of maize. I focus on the role of maize as
the primary production plant of the PPC. I describe how its food characteristics are central
to this role, and to placing the promise of the technology, as outlined in an interview with
Professor A.:

Professor A: Of course there's this big debate, I don't know if you've come across it,
about whether you should grow your, express your medical products in a food plant
or avoid it like the plague, I mean that‘s something that has come up
RM: What's your view?

192
Professor A: Well I mean its one of those awkward things because, in some ways,
obviously people choose edible plants because a) they're not toxic, they have no
toxins, and er, an awful lot is know about processing, because they're actually used.
(Professor A.)

The food characteristics of PMPs both support and threaten its promise. Professor A.
outlines how biopharming‘s products embody narratives of food production and
consumption. The safety of the pharmaceutical product is established and reinforced
through the characteristics of food plants themselves. Foods are ‗good‘ for biopharming as
they are non-toxic and contribute to the achievement of pharmaceutical safety without the
need for complex processing. The advantages of maize as a food production platform are
further outlined by Professor L.:

The reason why maize is technically better for me is that the protein is stored in the
seed, which is a tissue which is biologically developed for protein storage, so you get
tonnes of maize seed which is concentrated antibody and desiccated, and pre-
packaged for you if you like, and you can leave that in a silo for ten years if you like
before you process it. Whereas with tobacco leaves, it‘s a bit like the Birdseye peas,
you have to process it half an hour after you've cut it. So that‘s one advantage. The
second advantage is that the safety profile of maize is very well established, you know
we've been eating maize for centuries, whereas the safety profile of tobacco is a bit
more complicated. So in terms of downstream purification, the downstream
purification of maize should be very simple
(Professor L.)

The characteristics of maize constitute its advantages, both in terms of the ability to store
pharmaceutical products and in terms of its ‗safety profile‘. Maize is inherently stable and
safe. The second of these characteristics is established through what, in the language of EU
regulation, is termed a ‗history of safe use‘ (Constable et al. 2007). For researchers, the
safety profile of maize, based on its food use, contrasts with that of alternative
technological options. As introduced by Professor L., the PPC‘s alternative crop, tobacco,
is handicapped by its more complex safety profile and the need to process the product
quickly. As an unstable non-food crop which contains nicotine and toxic alkaloid
compounds (Schiermayer et al., 2004: 92) it is a ‗bad‘ crop for biopharming. The relation of
PMPs to edibility and the purity of food products relates the promise of biopharming to a

193
tradition of the safe consumption of traditional food crops. Discussion of purification is
used by researchers to introduce the advantages of the existing ‗purity‘ of plants,
particularly foods. Plant, particularly food crop, production is characterized as inherently
safer than other forms of biopharmaceutical production. The narrative placing of
biopharming‘s promise in relation to food is accompanied by a material relation to food
products. Concurrently, the safety of foods differentiates plants made from existing
pharmaceutical production methods, which carry the risk of contamination with pathogens.

Within the PPC, the hybrid nature of the technology is evidenced by the processing and
production methods used. At one laboratory, I was shown the domestic blenders and
kitchen technology used to initially process the pharmaceutical product. The role of
processing, or the lack of it, in the use of food crops plays a significant role in the
imaginaries of production mobilized by PMP researchers. The decreased requirement for
processing is taken to its conclusion in the example of edible vaccines, in which the
boundaries between food and medicine are blurred further. While the primary emphasis is
on maize, research into the production of subunit vaccines in lettuce and tomato is a
‗development loop‘ in the PPC. In an interview, Professor D., one of the senior researchers
involved in this development loop, describes the logic behind this research:

There are two schools of thought and essentially they are both in PharmaPlanta. One
is that you are using plants as factory to produce things that you will then purify, and
then and there are a group of us that believe why bother? Put it in something edible
and eat it.
(Professor D.)

‗Edible vaccines‘ formed part of the early promise of biopharming. For Professor A. they
represent a logical extension of the use of food crops to provide a low cost, low tech, stable
delivery system. The advantages of foods are not simply those that underpin the safety of
the production system, but are instead incorporated into the products themselves. As Ma et
al. (2003) continue, edible vaccines, particularly those produced in bananas support the
geographical promise of biopharming, as:

―they are widely grown in the countries in which vaccines are most needed and can
be consumed raw or as a puree by both adults and children‖
194
(Ma et al. 2003:78)

The characteristics of the food product contribute to establishing the promise of PMPs, and
in the development of alternative biopharmaceutical production. The adoption of particular
food crops is linked to the imagined geographies of global agriculture. However, many of
the appealing characteristics of edible vaccines give rise to their most significant socio-
technical challenges. In particular, the material relation of edible vaccines with food is
problematic, due to the breakdown of food in the digestive tract and the difficulties in
ensuring standardised dosing. The intimate combination of food and medical products
makes separating the response of patient bodies complex. This is outlined in a critical
feature in Nature in 2004, in which an interview with vaccinologist Stanley Plotkin
described the problem of 'tolerization':

―If vaccines are intimately presented together with food, the gut‘s immune system
faces a conundrum,‖ he notes. The gut is designed not to react to antigens in food,
but must produce a useful response against the vaccine. Instead of being immunized,
patients could even end up being ‗tolerized‘, meaning an immune response against
future invaders would be weakened, not intensified.‖
(Vermij 2004: 881)

The problems of tolerization, and of aligning food with pharmaceutical dosing and
replication requirements detract from the original edible vaccine model. However, this has
not resulted in a move away from foods entirely:

―it is important to recognize that edible vaccines will not be delivered as fresh
produce, as often suggested. A regulated product requires controlled delivery of
standardized doses, so some level of processing of the edible plant material would be
required. This need not include complex purification, but is more likely to involve
simple and inexpensive food processing techniques that are readily available‖
(Ma, Barros et al., 2005: 595)

Standardisation and control enables the qualities of food to become those of


pharmaceuticals, without a need to move beyond existing food technologies and expertise.
Food processing technologies bring the biopharmed product to a middle ground between
standardised pharmaceutical purity and unprocessed food. As depicted in Figure 5.1, this is

195
made material in a form which combines the stereotypical tablet form of pharmaceuticals
with the unmodified nature of the biopharmed plant.

Figure 5.1 Pills of crushed plant leaves (from Arntzen et al. 2005)

Expectations of PMPs derive from the low-tech, safe qualities of foods. However, the
example of edible vaccines highlights that too close an approximation between
pharmaceutical and food form is materially, or biologically, problematic. It suggests that
the model of ‗food‘ used by researchers is potentially underdeveloped. Furthermore, the
examples presented in Chapter One suggest that the ‗safety profile‘ of genetically modified
maize varieties is not directly inferable from traditions of maize consumption. Indeed, the
establishment of even conventional food safety and quality in everyday consumption is
complex, as described by focus group participants.

5.2 Consuming Foods

Focus group discussions of food highlight the role of publics as active consumers. They
demonstrate the complex ways in which participants define and assess the foods and
medicines they consume. Their complexity reveals the work that goes into establishing the
‗histories of safe consumption‘ described by biopharming researchers. For publics, both the
safety and quality of food products are continually re-established at the intersection of
personal assessment and more or less-trusted proxies and information.

An engagement with the superficial appearance and other sensory cues provided by the
materialities of foods is the first means through which this sorting of ‗good‘ and ‗bad‘ is

196
performed. In this extract from Group B (young urban female), Evie compares British food
to that in her native Greece, suggesting that:

Evie: Actually, food here seems quite industrial, it doesn‘t have any dirt or anything,
it‘s all very nicely packed, very tidy. I kind of, it doesn‘t look like real food, it looks
plastic
RM: Why is that?
Evie: Well in Greece you can find things in big coffins and they have all the dirt on
them, well they‘ve been cleaned a bit but you can sense, that they‘re more...
Jo: Of the earth
Evie: So it doesn‘t mean they‘re better, it‘s the look of it; it seems, here, it seems to
me more like plastic. But I‘ve got used to it now, but at the beginning I was, it was
very weird, the way the food is packed and given out. And it‘s so green, it‘s so perfect in a
way, the colour is so perfect
Jo: You mean like in the supermarkets and stuff?
Evie: Yeah, it looks unnatural to me, the lettuce is too green, the tomato is too red, it
hasn‘t it doesn‘t have any spots or anything
(Group B, meeting 1)

For Evie, the appearance of the foods establishes their authenticity. The dirt and
presentation of vegetables in Greece contrasts with the products of ―industrial‖ British
agriculture. Evie thinks that British food is plastic, too perfect and ‗unnatural‘, a caricature
of her normative image of food. As she elaborates, the authenticity of food involves a
connection with production which she sees as severed in the UK, what Jo describes as food
being ‗of the earth‘, or more natural. This builds on an earlier discussion among the same
group, in which they consider the foodstuffs provided as props. Initially, all participants
described how they would eat apples:

RM: So why so keen on the apples? What is it about them?


Evie: They're supposed to be natural and healthy
Rita: Unprocessed kind of thing
Evie: Thinking that hopefully they won't have that much kind of preservatives and
things in
Rita: Yeah, sprays and so on. maybe wax, but it‘s usually the red ones that they wax,
they do that to apples these days
Evie: They're all shiny, when you wash them…
Rita: You feel the substance, right
197
Sarah: Well the apples are kind of…they're a natural source as well aren't they, I
think it‘s one of those things as well isn't it
Evie: Sort of less processed, less…
Sarah: They've become associated with healthy living as well, there's that phrase, an
apple a day keeps the doctor away, so, it‘s kind of ingrained in you when you're
younger that you should have an apple. I do like the taste of them, but occasionally
I'll eat them because I have to
Jo: Because they're good for you
(Group B, meeting 1)

This discussion is at the start of Group B‘s first meeting, and highlights the intersection of
concerns about processing, healthy eating, price and the need for quick meals. The group
draws on two sets of characteristics about these products. Firstly, in the case of the apples,
they describe how they are, in theory, ‗natural‘, unprocessed and pure. When this is not the
case, as in the case of waxed apples, this can easily be identified by participants. Secondly,
they relate these characteristics of the apple to wider discourses and knowledges around
healthy eating and normative eating practices. The qualities of the foods alone would mean
nothing without the knowledge ‗ingrained in you‘ that apples are ‗good food‘. Such
concerns circulate widely in contemporary debates about food production, and both print
and television media are replete with discussions of healthy eating which are echoed in
group discussion.

However, as Patricia, a participant in Group E pointed out, while one might expect fruits
and vegetables to be healthy, they may in fact be ‗contaminated‘ or past their best. These
problems are exacerbated in foods whose matters are less open to scrutiny. Not all products
presented to groups as props, nor in wider circulation, are equally ‗transparent‘. For
example, among the props provided were probiotic yoghurts, intended to stimulate
discussion about healthy eating and uncertainty at the interstices of food and medicine.
However, these functional foods were regarded with some scepticism by focus group
participants, as described by Katie in the extract from Group F (young rural mixed).

Katie: I guess with the biscuits, you know they're not very good for you, you know
they're going to have lots of fat and sugar and things. But with the yoghurts you think
they might be good for you, but you don't actually know what’s in them, and you

198
hear lots of stuff about things that are meant to be good for you that aren't actually
as good for you as you thought
(Group F, meeting 1)

Katie‘s difficulty in assessing probiotic yoghurts derives from the absence of either visual
prompts or trusted information. Consequently, potential health benefits are outweighed by a
scepticism developed by analogy to other products whose promise failed to match the hype.
In contrast, although unhealthy, biscuits are at least certainly so.

Concern and mistrust of food quality and safety often emerges when their qualities are not
open to inspection, or are invisible, such as vitamin content. Consumer assessments of such
foods is supplemented by proxies such as quality assurance standards and food labelling.
Eden et al. (2008) describe how these allow consumers to identify qualities they can not
examine themselves. However, they shift the onus of proof from the product itself to the
institutions and bodies responsible for the provision of information. This can be seen in the
following extracts from Group E (older rural mixed):

Patricia: What I wonder about with the mashed potato I mean if you eat up
yesterday‘s mashed potato, it doesn‘t taste like it did day one does it, so what are they
putting into the commercial mashed potato?
Joan: Well you read the label don‘t you
Patricia: To make it still taste ok, so it can‘t be pure mashed potato can it? It must
have something in it
Joan: Well it tells you on the back what‘s in it
(Group E, meeting 1)

In this discussion, Joan counters Patricia‘s scepticism about processed and packaged foods
by referring to the labels and the lists of ingredients. However, as the group‘s discussion
develops, this labelling itself is questioned:

Helen: In foods I‘m a bit more cautious what‘s in the labels and what‘s on, what
they‘re made of, what additives are in there
Elizabeth: But there‘s so many to read, if you buy, when you‘re shopping if you look
at all the labels, it‘ll take you days
Helen: I know, my children think I‘m embarrassing, they go Mum, just put it in the
basket

199
Elizabeth: It‘s true, trying to find which like which country it‘s come from is very
difficult
Paul: No, but your labelling is never proper
Helen: I know, you want the labels to give you proper information
Paul: I mean old Bernard Matthews is English turkey, we know damn well it‘s not
(Group E, meeting 1)

Both the availability and limitations of labelling as an alternative to transparency are


opened up in this extract. In Helen‘s initial comment, the labels offer a means of
ascertaining what foods contain, acting as a reliable proxy. However, the perceived
usefulness of this information unravels through the discussion. Elizabeth and Helen discuss
the impracticality of reading large quantities of information in the context of shopping. Paul
and Patricia then suggest that the labelling itself is not reliable. The use of proxies such as
labelling, in making food choices relies on trust in those doing the labelling and in the
institutions providing the information. In this case, the example of the 2007 H5N1 avian
influenza outbreak at a Bernard Matthews turkey farm is used to reveal the topologies of
food production that had been ‗black-boxed‘ by labelling, and consider their relation to
food safety and quality (see also Donaldson and Wood 2007). This discussion introduces
the importance of the geographies of food production and of the spatial proxies adopted in
the absence of direct encounters with production.

Throughout the discussions above, food is discussed in terms of its quality, rather than its
safety, echoing the findings of Green et al. (2003) that safety does not represent the primary
means through which consumers engage with foodstuffs. Instead, food qualities are linked
to expectations of effects and outcomes of consumption. Participants especially, although
not exclusively, consider props as healthy or unhealthy, as in Group B‘s discussion
presented above. This is expanded in the extract below from Group C (older urban mixed):

Ian: Yeah, I'd say you've picked on products which are generally quite healthy, even
pasta itself is generally good, in theory anyway, you should be eating those
RM: What makes them things that are good to eat?
Catherine: Because it‘s fresh, fruits fresh and you know fruit‘s good for you
Violet: And it‘s got vitamins and minerals and...you know, it‘s good for you, fruit and
vegetables

200
(Group C, meeting 1)

The group consider the ensemble of props presented as discussion prompts. All six focus
groups are unanimous in this view of fruit and vegetables as appealing and healthy. The
specific qualities of food, such as the freshness of the fruit, contributes to placing the props
in relation to general characteristics of food. This is based on participants‘ own knowledges
of good foods. Violet, Ian and Catherine describe how you know that pasta, fruit and
vegetables are good for you in general. The individual, specific characteristics of foods are
accompanied by a large body of information about their generic qualities. In the extracts
above, the sources of this information are not specified – they are something that ‗you
know‘, or that is widely believed. Group F, the young rural group of teachers, bring the
work involved in the dissemination of food knowledges to the fore. This is both their own
work and that of media representations of healthy eating:

Josh: When I was growing up, I can't remember lots of adverts and things
recommending healthy food. The only thing I can remember is the Weetabix advert
with Ian Botham
Sam: No, not adverts, just TV. Documentaries and stuff.
[…]
Katie: Well when did the five fruits a day come in? That wasn't that long ago was it?
Rebecca: No
Alex: You can't go into a cover lesson today without it mentioning oh, make a
presentation on 5 a day
Katie: But that‘s something recent isn't it, I don't know if you've noticed it but you've
started seeing it in the supermarkets haven't you?
Alex: Yes, my Dad said that the other day
(Group F; meeting 1)

For Group F, healthiness is a relatively recent feature of the presentation of foods, and is
disseminated through a range of outlets – education, television documentaries, adverts and
in supermarkets. The value of eating five pieces of fruit or vegetables a day is constructed
through the work of teachers, supermarkets and the media.

The work involved in the construction of food advice is particularly apparent when groups
consider the relation of genetically modified (GM) foods to those provided as props. Here

201
the emphasis shifts from the quality of foods, to their safety. As described above, the safety
of foods is central to the technological promise of biopharming. However, as do food scares
around BSE or bovine growth hormones (Stassart and Whatmore 2003), GM problematises
‗safe consumption‘. It calls attention to consumer attempts to re-establish the safety of
foods which are concealed in the case of conventional foodstuffs, as described by Group A
below:

Rob: I wouldn't have a problem with GM crops if it was proven that there was no bad
effect that you'd get from eating them. There's this sort of scare that having something
that‘s genetically modified might cause, like create some sort of disease or have some
long-term effect on the body.
James: That‘s something that you can't really, you're not going to be able to prove
until long.., because you're going to have studies that say GM doesn't hurt you...there
are going to be studies that say they are
Will: Well you could one day have, prove it
James: But you're going to have to wait a long time
Anil: Yeah it'll be a long time
[…]
Will: It‘s, it‘s not particularly, well, you know, the analysis of whether it‘s working or
not. I sometimes scratch my head when I get a carton of Florida orange juice, and I
think, Florida orange juice - er United States, er modified food - and I, we don't
really know, and I'm sure we all at some point in time during this month will consume
some aspect of GM food just because...
James: I'm not denying that, but the whole idea is it‘s going to take time for people to
trust it, and you don't want to take the risk
(Group A, meeting 1)

While Will presses for the provision of reliable information derived from safety testing to
allow him to make an informed choice about genetically modified crops, James highlights
the uncertainty of such testing. He describes how tests are unable to resolve the question of
whether GM foods are safe or not in the short term. Instead, he suggests that it is necessary
for sceptics to place their trust in food production and to ‗take the risk‘ of GM. This view is
not based on the credibility of information about GM, but rather on James‘ own view of
technoscience which echoes the view of biomedical progress described in Chapter Four.

202
However, James‘ position is relatively isolated, and as the discussion develops, GM foods
are challenged further on the basis of their ‗authenticity‘:

Euan: Surely the whole attraction of food is that it‘s natural and that it‘s got nothing
done, it‘s got nothing wrong with it, like in the organic thing, the fact is that people
like it because it‘s natural and there's nothing wrong with it. If you start messing with
it, it becomes artificial and stops being natural
Mark: I think there is a fundamental fear of like messing with nature, and people
aren't comfortable with that idea
Will: I can see why
(Group A, meeting 1)

This discussion links the consideration of food safety, introduced above, back to the
qualities of foods. In particular, as did Group B‘s discussion of British food, it draws on a
distinction between ‗natural‘ and ‗unnatural‘ foods. It emphasises that the rejection of
genetically modified foods represents a continuation of the same behaviour as that for
conventional, but processed, products. Euan contrasts the ‗artificial‘ qualities of GM foods
with the ‗naturalness‘ of organic produce, linking these integral characteristics to the prior
discussion of safety. Group A‘s discussion re-emphasises the importance of ‗naturalness‘
and ‗reality‘ to consumer discussion of foods. These contribute to the assessment of
whether GM foods can be expected to be not only healthy, but safe.

Throughout these discussions, focus group participants are asked to perform as consumers,
making judgements about food safety and quality on the basis of what they themselves
would eat and then relate this to their own experience and knowledges. I explore these
knowledges through the examples of individual products, including those of biotechnology,
and how they are related to wider discourses and practices of food production and
consumption. Consumer discussion establishes foods as ‗good‘ or ‗bad‘, and ultimately as
‗safe‘ through their relation with ‗natural‘ or ‗real‘ alternatives. As expert discussion
describes, these classifications become reified in a ‗history of safe consumption‘. In the
process, the complexity of these discussions is subsumed within a homogenised
understanding of food safety.

203
5.3 Consuming Pharmaceuticals

The basiss on which quality and safety is established for pharmaceutical consumers
contrasts markedly with that of foods. As for foods, focus group discussion of
pharmaceuticals was prompted by the provision of props , consisting of
conventional/synthetic chemical pharmaceuticals and herbal or alternative medicines. The
conventional medicines included paracetamol or ibuprofen, aspirin and hayfever remedies.
The use of similar props allowed the same questions to be explored as in discussion of
food. In particular, it enables me to examine the behaviour of publics as consumers of
pharmaceutical products, and to contrast the informative materials of foods with the
reliance on information and trusted proxies in medicine.

While appearance and other material cues provide a basis for group discussion of foods,
such cues are rarer in the context of drugs. In one of very few group discussions
considering the appearance of medicines, Sofia describes her experience of pharmaceuticals
and introduces a distinction between drugs:

Sofia: You notice the difference in the tablets. Those I take for osteoporosis, I take one
a week, and they‘ve changed those. They‘re smaller and I get indigestion now, because I
haven‘t to eat anything for, or drink for so long after I‘ve taken it, and you haven‘t to lie
down
(Group D, meeting 1)

Sofia‘s regular encounters with medicines contribute to a commentary which is unique


among group discussions in its specificity. Sofia‘s description of her tablets for
osteoporosis echoes discussions of food choice. The appearance of the tablet is linked to
noticeable changes in effect. However, in other groups, where the material characteristics
of medical products are considered, it tends not to be in terms of individual products, but as
a generic pharmaceutical form, that of the tablet or pill, as in the extract from Group F
below:

Rebecca: I wouldn't swallow pills, but I trained myself with a bit of Mars bar, I
thought it was a bit silly people getting medicines and me not being able to, so I had
to learn. Can you imagine? Right, tonight I'm going to learn to swallow a tablet

204
Katie: I didn't use to swallow tablets until I was older either
Rebecca: It was when I had to have those soluble things and I had glandular fever
and I took some of them and it made me feel sick, so I didn't have any more of them
(Group F, meeting 1)

Group D‘s discussion of their tablets and Rebecca‘s comparison of pills and soluble
medicine introduces an important feature of pharmaceutical consumption that distinguishes
it from that of foods. While certain foods are seen as ‗unreal‘ on the basis of their
appearance, that of conventional medicines is generic and offers little guidance.

Rebecca describes how, in order to take medicines, she has adapted to be able to consume
tablets. In Group E, Jane makes a similar point in her relation to medicines, describing how
―I wouldn‘t take tablets because I can‘t swallow them‖. Pills are both less easily consumed
than foods, but also less avoidable in certain situations. While group participants describe
how they do not choose foods they dislike or can not palate, there is no choice but to adapt
to ‗take one‘s medicine‘. This contrasts with Marris et al.‘s (2001) description of choice in
pharmaceutical consumption – although in all groups participants describe how they try to
avoid medicines, only one participant in focus groups chose not to take any medicines in
any circumstances. ‗Opting out‘ entirely is the exception rather than the norm.

Because of the lack of information provided by the qualities of pharmaceuticals themselves,


there is much greater reliance on labelling and related information in attributing meanings
to pharmaceutical materialities. As such, examining how relations between pharmaceuticals
are established requires considering the role of the various actors involved in their
production and consumption. Information about the characteristics of pharmaceuticals is
provided on packaging and information leaflets, and by human intermediaries – doctors and
pharmacists. Confidence in medical information and doctors is highlighted in Group A‘s
discussion when they are prompted to compare their discussion of ‗natural‘ foods with that
of medicines:

RM: If I take some ibuprofen, that‘s not natural, why doesn‘t it matter there?
Rob: Because it‘s had stringent testing, it‘s been proven to work, it‘s, if, if you‘re, other
people have had it, and if it doesn‘t work, if it does cause you harm, then there‘s
someone that‘s accountable for it I suppose, and that you can pinpoint it down to the

205
aspirin, whereas with food like, you just don‘t know, it just feels so much less safe
because it just hasn‘t been through this same rigorous checks as medicine
Ed: I suppose again with medicine, you‘ve got the doctor as the one point of reference, that
we actually trust them, whereas with food you don‘t have that one person you can
trust
(Group A, meeting 1)

For Group A ‗naturalness‘ was an important feature of foods, particularly in contrasting


traditional with genetically modified varieties. In the case of medicines, this informative
contrast is replaced by the provision of reliable, tested information about effects and
accountability for this information. Trust in medicines is embodied in the doctors and
testing regimes. For Group A, both information, expertise and the traceability of medicines
is relatively unproblematic. For Rob and Ed, medicines approximate the ‗ideal type‘
described by Marris et al. (2001). However, as biopharming brings together not only the
products, but the institutions and actors of medicines and foods, maintaining this trust is
potentially difficult. Furthermore, in groups D and E, two of the older groups, in which
members appeared to have more experience of pharmaceutical consumption, a more
complex discussion of medical information took place. This can be seen as Hilda and the
other members of Group D respond to Sofia‘s description of her tablets above:

Hilda: I‘ve gone into this. I have to put eye-drops in, and there are two types, and
they‘re both exactly the same prescription, as to how it‘s made up and everything.
They try and con me in this, and I really do not agree with them, and I‘ve fought it
and it just depends, sometimes I get away with it, and sometimes I don‘t because the
chemist will say unless they‘ve put a certain thing on, you‘ll have to have these, the
cheaper ones, and I do disagree...
Sofia: I do think there‘s a difference in them
Hilda: There is
Sofia: And the doctor will say…I said ―is this the same?‖ and he says ―oh yes,
they‘re just the same‖, but they could be a cheaper you know…they do do that, I think
they do that
Hilda: Once you‘ve had experience from it, you know what you‘re… it‘s not hearsay,
it‘s happened to you.
(Group D, meeting 1)

206
For Group D, the appearance and visceral experiences of individual medicines are
introduced to problematise equivalence between pharmaceutical products, despite the
reassurance from chemists and doctors that the prescription is ‗exactly the same‘. The close
relation between product effects and trusted institutions described by Group A fragments.
However, here group participants cannot be the authoritative consumers they are in the
context of foods. In Group D‘s description, drug consumption and prescription becomes a
contest between expert and lay actors, in which each attempts to enforce their own
understanding of the identity of the pharmaceutical product. Hilda‘s endeavours to act as a
discerning consumer are thwarted by her chemist‘s definition of her medicine‘s relevant
qualities.

Information about medicines and their qualities is inseparable from the medical institutions
that transmit it. These institutions remain authoritative even when consumers attempt to
assert their own pharmaceutical knowledges. However, although Sofia describes her
firsthand assessments of her own prescription, and her belief that the doctor is not being
entirely honest with her, it is by no means clear that participants would be comfortable with
becoming more powerful consumers of pharmaceuticals. As the conversation in this group
develops, she describes how despite this, she is concerned about her ability to judge
pharmaceuticals. In the extract below, she explicitly contrasts pharmaceuticals and foods:

Sofia: You are, you‘re more frightened of going and buying something from a
chemist, I mean fruit‘s good for you, I don‘t care whether it‘s a Cox or what it is, it‘s
good for you. But there are so many different medicines now, you are frightened. I
think that‘s why I don‘t take tablets unless I‘m forced
(Group D, meeting 1)

For Sofia, choice based on the assessment of the specific qualities of individual products in
the same way as for foods is infeasible, except for those products with which she is
experienced. In discussion of medicines, the focus group participants in the extracts above
present themselves not as consumers of pharmaceuticals, but as patients for whom
medicines are provided. As such, the role of trusted institutions and authorities is more
evident, as Will expands on this in the extract below:

207
Will: I think the medicines that we actually are familiar with are very few compared
to those we actually can buy over the counter. We generally tend to buy these sort of
things because we are quite familiar with them, and for anything more than a bit of a
sore throat or a headache, or a cut, we'll go to the doctor and be prescribed.
(Group A, meeting 1)

In the younger group, Will‘s concerns are not about the medicines provided as props, but
about those for more serious conditions. He introduces a distinction between familiar and
unfamiliar products. The purchasing of the former is on the basis of experience. Beyond
that, the doctor acts as a reliable source of information and of products themselves.
Reported information is thus more important than in the case of foods. These discussions
focus on the role of doctors and chemists in providing the information. In the extract below,
Group E explore further the relationship between information about drugs and food, as I
prompt them to discuss how they classify ‗good medicines‘:

RM: How do you know what good medicines are as opposed to what good foods are?
Joan: Well in your medicine you have a leaflet that tells you all about it don‘t you in
the box. If you read all that it tells you the cause of the problems
Douglas: If you read all that you wouldn‘t take it!
[laughter]
Joan: But the object of the paper is to make you aware of the side effects
(Group E, meeting 1)

The characteristics of foods were used to classify them as ‗good‘ or ‗bad‘, including
placing them in relation to a discourse of healthy eating. The extract above introduces how
information about pharmaceuticals is used to classify them in relation to a similar focus on
the effects and side effects of consumption. Whether pharmaceuticals act as intended forms
a central part of participants‘ discussion and allows Group E to determine ‗good
medicines‘.

Discussion of conventional medicines classifies them as ‗good‘ or ‗bad‘ on the basis of


their expected positive (deliberate) and negative (side) effects. However, information about
these effects is not itself seen as certain. Although trusted, medical advice and knowledge is
seen as partial and subject to change, as described by Helen in Group E:

208
Helen: When I was a lot younger… it was hayfever I had tablets for, and I just had
them on repeat prescription, and they did do the trick, when I was you know, snuffled
up. I took these things and they did the trick, but then I went back to the doctor‘s one
day to get my repeat prescription and they just said you can‘t have it any more, you‘ll
have to go back to your doctor and see what they can do, and I said why, and they
said they‘ve been withdrawn because of the side effects. You can imagine at the age I
was, and how long I‘d taken them, I was horrified, I was thinking you know, what‘s
going to happen to me? What‘ve they done to me?
Jane: But there are drugs that have horrific…
Helen: And it put me off taking things, until then I hadn‘t really thought about it you
know
(Group E, meeting 1)

Helen‘s description of pharmaceuticals is ostensibly critical of the uncertainty of


pharmaceutical effects, and has deterred her from consuming pharmaceuticals. However, as
described in Chapter One, Marris et al. (2001) suggest that the ability to trace, identify and
withdraw damaging medicines from the market represents a key point at which they are
distinguished from foods. Group A‘s discussion above emphasises this specificity of
effects. In the case of biopharming, this ability to trace and identify pharmaceuticals
produced in food crops may be diminished.

5.3.1 Natural Medicines

Along with conventional agriculture and pharmaceuticals, biopharming intersects with the
products of existing herbal or botanical medicines, although the relevance of this link is
contested by both publics and experts. However, the herbal medicine products provided as
props are the focus of a public discussion of medicines in which focus group participants
adopt a more active role to that described in the previous section. For herbal medicines, a
discourse can be seen that draws on elements of those around both food and medicines. In
particular, participants reintroduce the role of ‗naturalness‘, important in discussion of
foods, but less so in considering conventional pharmaceuticals.

In the case of herbal medicines, focus group discussion is dominated by the material
characteristics of products and accounts based on personal experience. These relate more to
food knowledges than to the pharmaceutical ones described above. However, more
209
negative assessments of herbal medicines contrast them with conventional pharmaceutical
products, emphasising the lack of reliable information and bringing the role of testing to the
fore.

Figure 5.2 St John's Wort in tablet form

The props comprised a series of herbal medicines, including arnica ointment, a box of St.
John‘s Wort tablets, as shown in Figure 5.2, and tea tree oil. Jo in Group B (young urban
female) uses the appearance of these products to introduce distinctions between ‗natural‘
and ‗processed‘ herbal and plant remedies:

Jo: I think these all look like they were produced in a factory, very sterile, not
particularly natural, whereas like garlic or lemon might not be all that natural with
all the pesticides that have gone into it, but it seems a lot more organic to put into your
body, you kind of know what you‘re getting, but you know a kind of pill, it could be
anything, its kind of, like the shape that I recognise I suppose
(Group B, meeting 1)

Jo contrasts the sterility of factory produced herbal medicines and the more ‗natural‘ form
of medicinal foods and herbal medicines represented by garlic or lemon. The appearance of
the products is important in delineating between industrial and natural medicines. As Jo
considers herbal medicines, the distinction between natural and unnatural forms is used to
highlight uncertainty about the contents of processed products.

210
In an echo of discussion of foods, ‗naturalness‘ is used to indicate authenticity. Jo
introduces the importance of appearance and naturalness in distinguishing between
‗conventional‘ and herbal medicines and in establishing typologies of the latter. I have
suggested that information about the positive and negative effects of pharmaceuticals is
important in classifying ‗good‘ medicines. As groups consider herbal medicines,
‗naturalness‘ is described not just in terms of the authenticity of the products, but as related
to the characteristics of the drugs discussed in the previous section. In Group C, Catherine
contrasts the side effects of conventional medicines with her experiences of herbal
medicines:

Catherine: The side effects of these drugs, sometimes they‘re not always known are
they? Like you were saying Patience, about using the actual plant. When I was in the
Caribbean, in Barbados, we had stuff called aloe vera, and it was the most
amazing…I got sunburn, and I used this aloe vera and it was absolute…and this is all
natural
(Group C, meeting 1)

For Catherine the risk of side effects is related to the ‗naturalness‘ of the product. In contrast,
for participants in Group A, the effects of medicines were comprehensively described by
doctors and characterised by testing. In place of the information provided by medicine and
doctors, Catherine uses ‗naturalness‘ to describe the efficacy and lack of side effects of
botanical medicines. Calnan and Williams (1992) have argued that a turn to complementary
and alternative medicines is part of an estrangement from biomedical interventions and a
return to natural and perceived 'safer' forms of healing. More recently, Calnan et al. (2005)
describe how complementary and alternative treatments are seen as less harmful than
conventional pharmaceuticals because they are ‗natural‘.

In section 5.2., I described Katie‘s scepticism about probiotic yoghurts as ‗healthy‘ foods.
In Group E participants draw on their own experience, the product‘s appearance and
second-hand information in discussing the same prop in the context of herbal medicines:

Elizabeth: We have the tablet, the Acidophilus, which is what‘s in those,


Jane: That‘s right

211
Elizabeth: But those don‘t get down enough to do anything, whereas a tablet will get
right down
Patricia: They‘re not pure enough are they
Elizabeth: No, no
Helen: Expensive aren‘t they?
Jane: Would you not get the same effect from eating a good yoghurt?
Joan: I would think so, yeah
Elizabeth: But there are different strains of bacteria that get further down into the gut
and…
John: Theoretically they do

Elizabeth: We went to a herbalist and she actually said to try this because Douglas
was suffering a lot with your nose weren‘t you, with catarrh,
Jane: And have you felt the benefit?
Douglas: Yeah, I can breathe through my nose now
Patricia: I might try that for my husband then
(Group E, meeting 1)

There is initial scepticism about the effects of probiotic yoghurts. In this group however,
participants in this group can testify persuasively to the efficacy of the probiotic. Moreover,
they do so through engaging with the material form in which the Acidophilus is presented
to the group. The yoghurts are seen as ineffective and insufficiently pure in comparison
with tablets. The presentation of the same bacteria in a concentrated tablet form allows it to
move from sceptically received foodstuff to professionally attested medicine. Rather than
being comparable to ‗a good yoghurt‘, the provision of a pill from a trusted source relates
probiotics to pharmaceuticals. Moreover, the redefinition of the product in terms of the
Acidophilus bacteria reduces the complexity of the food product to a simple active
pharmaceutical ingredient. The ‗purity‘ of pharmaceuticals is here linked to pharmaceutical
materialities, rather than the qualities of foods.

In these discussions, the ‗reality‘ of herbal medicines was related to its naturalness, while in
Group D‘s conversation the efficacy of the product as it moves between food and medicine
is bound up with the tablet form. Pills and tablets play a dominant role in focus group
debate. As generic pharmaceutical form they stand in for differentiated active
pharmaceutical ingredients. However, they also contribute to establishing pharmaceutical
212
typologies and sorting industrial, chemical or ‗conventional‘ pharmaceuticals from herbal
and homeopathic medicines and foods. They form a material point of comparison between
the promising futures of pharmaceutical research and the doubtful potential of herbal
medicines described in Chapter Four.

However, where the experiential testimony provided by Elizabeth and Douglas in Group E
is absent, discussion of herbal medicines is less positive, as in group A:

Euan: I've still got an aversion to herbal remedies, I don't know why, but you know I
read things that say it‘s all a lie.
Will: But don't things like aspirin start off as a herbal remedy really
Euan: No, I'm sure they do, I just look at this [arnica] and I see it as a bit of a
gimmick
Ed: Yeah, herbal stuff does have a gimmicky feel to it
Euan: That‘s how I react to it anyway
Will: But...dockleaves
Euan: Yeah, I know it‘s probably illogical, I mean a lot of medicines come from a
sort of natural thing I know, but when something calls itself...you wonder why it‘s not
calling itself a medicine, why it‘s calling itself a herbal medicine
(Group A, meeting 1)

As does the expert discourse around PMPs and botanical medicines in Chapter Four, Will
highlights genealogical links between aspirin and herbal remedies. However here
‗naturalness‘ and the form of the medicines contributes to their ‗gimmicky feel‘. For Euan
herbal medicines are ineffective by virtue of their self-differentiation from other medicines.
Similarly, Sam in group F, suggests that ―if they did work, they'd be more mainstream‖.
Rather than acting as a ‗commemorative moment‘ that reinforces the potential of herbal
medicines, as described in Chapter Four, here the discovery of aspirin only highlights the
difference between ‗real‘ and ‗gimmicky‘ medicines, between those which work and those
which only purport to work.

In the preceding sections, I have described how publics ‗place‘ existing foods and
pharmaceuticals through differing engagements with their material qualities. These provide
the basis for considering public discussion of biopharming. They highlight the role of the

213
aesthetic and material characteristics of food, pharmaceutical and herbal products in the
development of expectations of their efficacy, healthiness or safety.

5.4 Experts: The Problem of Pharmaceutical Equivalence

The products of novel technoscience not only embody its future, but are the means through
which this future is encountered and interrogated. In the extract below, Dr I. suggests that
the products of biopharming cement the associations with pharmaceuticals and
dissociations from agriculture required to mobilise public support for biopharming:

―The feedback we've got is that people are much more likely to support medicines
from plants than they are food from GM plants, possibly because plants are used to
produce medicines anyway so people are used to the fact that their medicine comes
from plants, and the fact that what they get in the bottle at the end is not going to be
half a tree, but is going to be you know a clear liquid. It’s no different really from
obtaining it from bacteria‖
(Dr I.; emphasis added)

Although he does not describe where ‗feedback‘ is coming from, for Dr I. the equivalence
of PMPs to existing biopharmaceutical products emphasises the inability of publics to
distinguish between pharmaceuticals, and establishes the place of biopharming within a
pharmaceutical tradition . His argument is that the production of pharmaceuticals in plants
is no different from production in CHO cells or bacteria and that the identical form of the
pharmaceutical product cements this correspondence. However, as I consider below, the
establishment of material equivalence between plant made and existing pharmaceuticals is
not quite as simple as Dr I. suggests.

For the production of biological medicines, ‗the process is the product‘. Establishing the
equivalence of one pharmaceutical to another therefore relies on occluding differences
between production processes in the final product. Indeed, biopharming researchers
describe how a key step in the development of biopharming is to establish the similarity
between its products and those of other production methods (Sparrow et al. 2007). As such,
PMPs must be brought in line with existing regulations which emphasise the importance of
safety, efficacy and quality, and guarantee the ‗identity, potency or efficacy‘ of biological
214
medicines (EC 2008). Regulatory approval provides the end point for pharmaceutical
products and thus the starting point to understand the characteristics of pharmaceutical
products.

However, establishing equivalence for biological medicines is particularly complex.


Chemical drugs have a structure that is nominally determined by their chemical formula,
yet even here, diverse and locally situated understandings of equivalence and similarity
circulate such that what counts as both ―sameness‖ and being a ―copy‖ is remarkably plural
(Hayden 2007). In contrast, the complex structure of proteins is never fully reproducible.
Protein drugs are inherently more variable than those produced by chemical synthesis, and
"even under the most stringently controlled culture conditions, proteins show a high degree
of heterogeneity" (Schellekens 2004:1357). Hence the 2008 EMEA Guidelines for
biopharming attempt to outline conditions of equivalence for plant derived proteins:

―Characterization studies should include a comparison of the product with its natural
counterpart, when feasible and relevant. The potential impact of the differences observed
should be carefully considered, and thoroughly discussed with regards to safety and
efficacy‖.
(EMEA 2008:8)

There are potentially problematic differences between proteins produced in plants and their
‗natural counterparts‘. I discussed earlier the role of ‗naturalness‘ as providing a basis for
the classification of foods in focus group discussion. Here, naturalness similarly provides a
baseline for comparison of novel biopharmaceutical products.

The analyses EMEA request include the determination of the overall monosaccharide
composition and the oligosaccharides released from, and attached to, the protein. In
addition, characterisation studies are to include analysis of post-translational modifications
(EMEA 2008). This analysis constitutes the protein molecule as a key locus in the
establishment of equivalence between plant based and existing biopharmaceuticals. The
majority of mammalian proteins undergo some form of modification during or after their
synthesis. These include the addition of extra chemical groups through phosphorylation,
carboxylation, hydroxylation, and most commonly glycosylation, the addition of glycan
chains. The latter forms the focus of much regulatory and researcher discussion.
215
Glycans are simple sugars added as an assembled protein passes through the endoplasmic
reticulum and Golgi apparatus of the cell, and which contribute to the folding of complex
protein structures, particularly those of monoclonal antibodies. The ways in which the
structure of these sugar chains is constructed differs between plants and other systems. In
particular, plants do not add sialic acid to the end of glycan chains, while the chains may
contain fucose and/or xylose residues. In the EMEA guidelines, these differences in
structure are taken as potential indicators of a difference in action and effect:

―A comprehensive characterisation of the plant glycosylation pattern, both


qualitatively and quantitatively, should be provided. This analysis should include the
determination of the overall monosaccharide composition, the analysis of
oligosaccharides released from the protein (e.g. determination of antennary
structures, mapping) and oligosaccharides attached to the protein (e.g. glycosylation
per site, glycoform distribution)‖
(EMEA 2008: 8, my emphasis)

Glycosylation provides potentially significant differences between plant made and other
pharmaceuticals. This is reflected in researcher publications. In Twyman et al.‘s review of
biopharming for Expert Opinions on Emerging Drugs, they suggest the importance of
glycosylation ‗cannot be understated‘ in attempting to achieve a consistent pharmaceutical
product (Twyman et al. 2005: 4). Others emphasise the potential consequences of altered
glycosylation for the immunogenicity of plant derived antibodies and raise the possibility
that plant glycans may be allergenic (Faye et al. 2005; Gomord and Faye 2004; Gomord et
al. 2004, 2005). For these authors, the requirement for glycosylation becomes central to the
success of biopharming in equating the products of existing and established
biopharmaceutical processes:

―more than one-third of approved biopharmaceuticals are glycoproteins and both


their function and efficiency are affected by the presence and composition of their N-
glycans … This absolute requirement for glycosylation explains why many
biopharmaceuticals are produced in expression systems with N-glycosylation
capability and, particularly, why mammalian cell lines are currently the preferred
host for therapeutic protein production‖
(Saint-Jore-Dupas, Faye, and Gomord 2007:317, my emphasis)

216
Here the glycan structure of the protein is inseparable from the function of the
pharmaceutical protein and consequently the viability of the plant production platform for
pharmaceuticals. This is an ‗absolute requirement‘ and represents a major point of
divergence between plant and mammalian biopharmaceutical production systems. This
position is developed in interviews, in which researchers echo the language of the
regulatory documents in describing glycosylation as important in terms of equating
‗naturalness‘:

RM To come back to glycosylation, do you think that at some point humanising the
proteins will have to happen?
Professor A: It probably will, I mean it‘s probably a good idea, you know to do it,
because it may be I suppose the more similar you are to the natural product, the
better it is. I mean even if it‘s only in terms of half-life in the blood stream that can
make a difference. As I say, if you‘re looking just for straight binding, 99%, it
probably doesn‘t matter. But for you know, neutralization, binding of complement and
things for processing it always will be a good idea to make it as human as possible, as
humanly possible.
RM Beyond functionally equivalent then?
Professor A: Yes, well yes, that would presumably be the ultimate, if you make them
look identical, you know
(Professor A.)

While the purpose of similarity is deemed to be 99% symbolic, a question of the


presentation of the medicinal product which does not affect the binding of the proteins, it is
still described as a ‗good idea‘. The role of glycosylation in establishing the equivalence of
PMPs is framed in visual terms – the aim is to ‗look identical‘ to natural counterparts.

The first response of Professor A. introduces uncertainty surrounding glycosylation. While


the papers by Gomord and Twyman suggest that equating mammalian glycan patterns is
important, Professor A. limits himself to commenting that it is ‗probably‘ a good idea. In
other interviews, discussion of the importance of glycosylation is characterised by
uncertainty. In the extract below, Dr G. describes the relationship between biopharmed
proteins and ‗natural‘ equivalents as a ‗philosophical problem‘ rather than one of any
material relevance:

217
It [glycosylation] is very controversial, because there are people who say plant
glycans are allergenic so by definition so you shouldn't make proteins in plants, and
then there are the enlightened [laughs] people who say well we've been eating plants
since we were kids and of course we've got antibodies against plant glycans, so who
cares? As long as the molecule does its job and it does it exactly like the mammalian
counterpart, who cares if its got a couple of xylose residues?... It is a philosophical
problem really, ok it‘s true that the molecules not 100% the same, but it depends on
what you want to do with it
(Dr G.)

Reactions to plant glycans are, for this researcher, known and certain, given the history of
consumption of plants as food. The different application to which these plants are directed
is essentially irrelevant; instead it is the functionality of the molecule that will determine
the medicinal success of biopharming. Despite concern about glycosylation patterns, Dr G.
suggests that it is efficacy, rather than appearance, that will result in the classification of
biopharming as pharmaceutical. Dr G.‘s position represents a continuation of the approach
to biopharming described in the previous section, in which food characteristics underpins
the promise of biopharming. This is made material in the form of plant glycans, and is
expanded in an interview with Professor B.:

There's two arguments, is the problem of humanising totally artificial because the
regulators want all drugs to be the same, or is the problem of having plant glycans on
a human protein will cause adverse reactions in the patient. And as far as I'm aware
a lot of people think that they won't have any adverse reactions in the patient because
our immune system has been blasted with them for ever, throughout the whole of
evolution.
(Professor B.)

Professor B. distinguishes between glycosylation as an ‗artificial‘ or a ‗real‘ problem to


suggest that the desire for equivalence is related to inflexible regulatory demands.
However, in a ‗top-down view‘ of biopharming from the pharmaceutical industry, the
importance of the appearance of pharmaceutical molecules is described by Freidrich
Bischoff of Boehringer Ingelheim, one of the world‘s 20 largest pharmaceutical
manufacturers (EFPIA 2008):

218
―[I]t seems that differences in glycosylation have little impact on the immunogenicity
of plant derived vaccines. Their efficacy has also been demonstrated in clinical tests.
Nevertheless, the pharmaceutical industry would prefer unchanged glycosylation
patterns in order to avoid possible side effects caused by nonhuman glycan
structures.‖
(Bischoff 2004: 274)

While Bischoff accepts that the safety and efficacy of plant derived pharmaceuticals, in this
case vaccines, has been demonstrated, from a pharmaceutical industry perspective, this is
insufficient. Whether or not glycosylation is biologically important is, in the short term
development of biopharming, essentially irrelevant, but nevertheless represents a
significant material restriction of biopharming‘s future. For Bischoff, as for pharmaceutical
regulators, any form of structural variation is seen as indication of a potential side effect.
Data about side effects and efficacy is thus secondary to achieving material equivalence
and ‗unchanged glycosylation patterns‘. Consequently, it is unlikely that these actors would
be satisfied by the use of maize to produce pharmaceuticals without the ‗humanisation‘ of
the glycosylation pathway.

For experts, biopharming‘s products are an important site in establishing presentational and
functional relations between plant made and existing pharmaceuticals and foods. Even
though researchers are familiar with the actual form of PMPs and their similarities with
other pharmaceutical proteins, the importance of material differences between these is
uncertain.

For EMEA regulators and pharmaceutical manufacturers the structural similarity of


pharmaceutical proteins to ‗natural‘ counterparts allows them to be placed in relation to
existing pharmaceuticals. The discussion of glycosylation highlights a division between
differing types of equivalence: functional, in terms of the effects of pharmaceutical
proteins, and structural, in terms of the form of these proteins. Achieving the appearance of
human proteins would ensure presentational and functional characteristics of
pharmaceuticals were met.

For some researchers, the negative effects of glycosylation are less certain. Instead,
glycosylation is part of the food qualities of PMPs. The purity and safety of food products
219
is a central part of the promise of biopharming, separating it from riskier animal production
platforms. However, as described in the case of edible vaccines, too close an approximation
between pharmaceutical and food products can be problematic.

5.5 Publics: Troubled Consumption

Members of the public use a range of qualities of foods and pharmaceuticals to classify
them according to their effects. Public discussion of biopharming discursively and
materially relates PMPs to these existing products. I now examine how the artefacts of
existing, ‗mundane‘ technoscience become mobilised as resources in the classificatory
placing of biopharming and its future.

Tablets, particularly paracetamol, anchor discussions of existing medical products. In the


extract from Group B (young urban female) below, Jo describes molecular farming in these
terms as part of a sceptical perspective on its potential safety:

Evie: It‘s one thing to have plants that already exist, and take their qualities into
medication or whatever or food, and another thing to take plants that already exist
and modify them, to me.
Jo: A bit Charlie and the Chocolate Factory-ish
RM: What did you mean by Charlie and the Chocolate Factory?
Jo: That well, I was just thinking about his sweet that he develops that, you know, you
put it in your mouth and it gives you a three course dinner, and I‘ve just got this
vision of you know a flower with a paracetamol pill kind of inside, and you know, you
take it out, and I mean obviously it‘s not as simple as that but you know, maybe that‘s
wonderful, I don‘t know,
Sarah: But then in Charlie and the Chocolate Factory she turns blue
Jo: Well that‘s what I was thinking, I was thinking that, he hadn‘t quite got it right
and she ended up being purple for the rest of her life, and you know he didn‘t know
that was going to happen till she did it, so...
(Group B, meeting 2)

The image of a plant containing a pill or tablet illustrates the uncertain consequences of
innovation and establishes biopharming‘s relation to existing medicines and their
unpredictable futures. When Evie distinguishes biopharming from existing plant medicines,

220
Jo draws on a fictional example to highlight uncertainty. She uses the fate of Violet Beau-
regard in Roald Dahl‘s Charlie and the Chocolate Factory (Dahl 1967), who falls victim to
the adverse effects of an untested product, to illustrate concern about PMPs. The superficial
simplicity of ‗a pill in a flower‘ is complicated by the problems of predicting and knowing
their effects.

In discussion of herbal medicines, Jo introduced a distinction between ‗natural‘ and


‗processed‘ varieties, suggesting that ―you know a kind of pill, it could be anything‖. Here
biopharming is defined in terms of processed pharmaceuticals. Evie also describes how
biopharming involves changing the existing characteristics of plants, separating
biopharming from herbal medicines, in a comment that parallels the relationship between
GM and conventional foods.

Group B are not alone in describing the products of biopharming as tablets or pills. For
example, Group D (older rural women) assume that PMPs will be tablets similar to those
they already take, while James in Group A (young urban male) assumes that biopharming
will involve ―new forms of tablet‖. As described above, the form of medicines is important
to groups both in ‗sorting out‘ conventional and herbal remedies. A similar pill-based
aesthetic linking biopharming to paracetamol is evoked in other group discussions. Group F
(young rural mixed), considers PMPs in similar terms to Group B.

Rebecca: But when they extract it, surely they do something with it, so therefore it‘s
quite chemically...
Max: Refined or processed
Katie: You mean it doesn't come out in tablet form
Rebecca: In the maize!
Katie: Instead of little bits of corn, you'd have little paracetamols
Rebecca: So you'd still be taking a tablet
(Group F, meeting 2)

Katie and Rebecca expand their discussion through imagined products of biopharming,
drawing on similar imagery to that in Group B. The image of tablets instead of maize
kernels is used to reinforce perceived similarity between plant-made and conventional
medicines. Throughout the two discussions, biopharming is associated with conventional

221
pharmaceuticals such as paracetamol on the basis of their imagined form as a tablet.
Placing biopharming in pharmaceutical terms distances it from group discussion of foods.
This is reinforced through the emphasis on the processing required in pharmaceutical
production, as the products of biopharming are not ‗little bits of corn‘.

In these extracts, the imagery of biopharming is that of processed conventional


pharmaceutical products extracted from a food or agricultural host. However, the medicines
remain separate and distinguishable from the food characteristics of this host. In these
images of biopharming, the form that they take suggests that despite the combination of
agricultural and pharmaceutical production, the products of each remain separate. To
further investigate the role of the product in placing biopharming, it is worth considering
the role of specific examples in group discussion. Together with the examples in the
information booklet, the idea of ‗edible vaccines‘ was introduced. Edible vaccines are not a
core target of the PPC; however, they are a ‗development loop‘ within the consortium.
More importantly, they are significant in the history and imagery of biopharming and recur
in popular representations of biopharming (for example in Vallely 2009).

The example of edible vaccines represents a point at which group discussion often departs
from a generally positive conversation about the promise of biopharming. In particular, the
mixing of pharmaceutical and food qualities such that they are indiscernible raises different
questions for participants from PMPs which maintain their pharmaceutical form, as
described by Peter from Group C:

Peter: One thing I can see…The public reacted quite badly to GM crops, what I can
see is people who don‘t need it for these conditions not going for it at all. I can see
many people with serious conditions saying we‘ll go for this, if it‘s easier than
sticking needles in us. But I think if there‘s, like a kind of a potato which is like a
paracetamol, I can see people not really using that for general use. I think there‘s a
suspicion in people.
(Group C, meeting 2)

The risk of contamination and the difficulty of knowing the properties of foods is
commented on in discussions of edible vaccines, and can be related to similar discussions
in the first group meetings. Peter introduces a distinction between publics, suggesting that

222
while food consumers might be suspicious, those acting as patients may not. However, in
the extract from Group F below, discussion of edible vaccines emphasises that these roles
can not be so easily split:

Rebecca: So would they, as a vaccine, would they then extract it


RM: The ones in [the booklet] as examples would be extracted
Rebecca: You wouldn‘t just eat your corn on the cob
RM: well one of the original ideas was to put a vaccine in a banana and have it as
something you could eat
Rebecca: See that, I‘d like that, that‘s clever
RM: You think it‘s a better idea
Max: Good way of getting it into kids isn‘t it, well not in a banana maybe...
Katie: It‘s like the polio on the sugar cube, and I had it and thought it was a reward
for being good for my injections, I never realized it had the vaccination on it
(Group F, meeting 2)

This group have already distinguished PMPs from food, and this is emphasised in
Rebecca‘s statement that the final pharmaceutical form would not be ‗a corn on the cob‘.
The example of edible vaccines is introduced to further examine this separation between
foods and medicines. Unlike for Group D, the resultant discussion repositions edible
vaccines alongside existing vaccination strategies. For Group F, we are all patients at some
point, and avoiding injections is of benefit to all, not just those with chronic conditions. For
Max, the presence of the vaccine in bananas makes it potentially easier for childhood
vaccination, presumably as needles and injections could be avoided. Similarly, Rebecca
sees this plan as ‗clever‘, while Katie links it to the use of sugar cubes in polio vaccination,
an example which was also used to place edible vaccines in groups D and E.

The example of polio vaccination introduces a link between existing ways in which
medicines are administered and PMPs. This is used to underpin support for molecular
farming. However, a distinction is maintained between the idea of vaccines in bananas and
that of vaccines on sugar cubes. In the latter example, the substance of medicine and food is
neither ‗blended‘ (and inseparable) nor consumed in the same way as imagined for edible
vaccines, but instead medicine is added to food within a medical setting.

223
While Group F‘s discussion introduces this relationship with polio vaccinations as positive,
other groups are more sceptical. The uncertainty about the content of foods discussed
earlier is reflected in further discussion of edible vaccines, as in Group E, in which edible
vaccines were again introduced by the facilitator:

RM: One of the original ideas for this research was having bananas that you could
eat, which would have the vaccine in them, so the vaccine would be in the food
Paul: Same as taking polio vaccine
Patricia: sugar lumps
Jane: Well that‘s what got me thinking
Helen: Would that be better, would it be better if it wasn‘t injected, if it was eaten
rather than injected?
Joan: At least an injection you know you‘ve had it
Jane: But as long as it gets into the blood stream
Paul: Yeah but it‘s, if it was done through food, you could well sort of overdose
couldn‘t you
(Group E, meeting 2)

Again, the production of edible vaccines is compared to the example of the polio vaccine.
However, this medical description is rendered problematic when a number of further issues
related to the ‗foodiness‘ of edible vaccines are raised. While Katie, in the extract from
Group F above, is unconcerned that her polio vaccination went unnoticed, in Group E a
distance is maintained between injection and ingestion. When they are contrasted by Helen,
Joan highlights the importance of the experience of injections in asserting control over the
administration of vaccines, so that ‗you know you‘ve had it‘. In contrast, ingestion is an
inherently unreliable means of delivery. Group E‘s discussion parallels concerns raised in
expert discourse of surreptitious administration and of ‗overdosing‘.

The first concern, that of surreptitious administration, reintroduces worries about


processing and interference with the ‗natural‘ or ‗real‘ qualities of food . Concerns about
unintended or clandestine exposure to vaccines are also raised in Group C, where mistrust
of doctors was strongest, particularly for Catherine. Again, discussion in this excerpt is
prompted by the introduction by the facilitator of the edible vaccine concept.:

224
RM: I mean one of the ways this research started was the idea of producing edible
vaccines in a banana or something. Would that, would people think that was a good
idea?
Ian: in theory it is a good idea
Patience: Yes, it‘s a good idea if the banana was produced without too many GM or
those things they use to spray the plants.
Catherine: Who knows whether we are being used as guinea pigs without our
knowledge? Who knows whether we are being pumped full of I don‘t know what
without any of us ever knowing about it? It‘s a possibility isn‘t it? Hasn‘t that been
done before, years ago when people were told to take something and they didn‘t know
exactly what it was, and they were being used as guinea pigs for certain types of
experiment and they didn‘t even know?
(Group C, meeting 2)

Patience situates molecular farming in relation to concerns about food production and
quality. However, Catherine considers it in relation to her broader distrust of institutional
medicine and the potential for people to be used as unwitting ‗guinea pigs‘. The provision
of pharmaceuticals in the form of food challenges participants‘ ability to control what they
eat and to judge and assess food. It draws on concerns about the ability of participants to
assert control over their own consumption of foods and pharmaceuticals.

Public discussion of biopharming‘s products initially places them among the pills and
tablets of conventional pharmaceutical production. Biopharming as medicine is generally
discussed enthusiastically. However, when the example of edible vaccines is introduced,
similarities between biopharming and food become of concern. Unlike for experts, the
similarity of biopharming‘s products to those of foods is not a desirable characteristic.
PMPs become an extension of existing uncertainty about the contents of food, but also
reveal challenges to the trustworthiness of pharmaceutical institutions.

5.6 Re-establishing Distinctions between Pharmaceuticals from


Food Products?

The central role of maize in genetically modified agriculture is extended in the case of
biopharming. The maize kernels of the PPC underpin biopharming‘s promise, providing a

225
means of producing medicines which are safe, stable and can be stored over a period of
years: a silo within a silo. For experts, this promise is extended in the use of other food
crops for the production of ‗edible vaccines‘, which build a new type of medicine on the
qualities of food. However, their promise is challenged by the difficulties in ensuring that
the product acts as a pharmaceutical rather than a food when consumed, and in ensuring
that small doses of vaccines do not ‗tolerise‘ the immune system.

For publics, the mixing of food and pharmaceutical qualities also raises serious problems of
separating foods and pharmaceuticals. In particular, the example of edible vaccines
introduces the necessity of developing, producing and supplying PMPs in manner that
protects the food chain from unintended exposure. In this section, I explore how making the
previously invisible spaces of transgene expression visible opens them to the material
discrimination between products. This is strikingly illustrated in figure 5.3.

Figure 5.3 Antibody producing maize kernels tagged with DsRed viewed under
normal (A&B) and UV light (C&D). Red carry the antibody, green do not. (from
Ma et al., 2005)

226
This picture was introduced to me by Professor L. as follows:

I've got to show you this, this is beautiful [shows picture of maize on computer]. So in
the PPC, what we've done is alongside the antibody molecules, we've inserted a
fluorescent marker so we can identify which seeds are transgenic. So there's the
maize crop, looks pretty good, and then you turn out the lights and shine a
fluorescent light on it and every red kernel has got the antibody in.
(Professor L.)

When the gene coding for the expression of antibodies is inserted into maize varieties, it is
‗tagged‘ with a fluorescent protein known as DsRed. The use of DsRed allows
contamination to be visibly detected. In images C and D, taken under UV light, red kernels
carry the gene, and are 'pharmaceutical' and green ones are non-transgenic, unmodified
‗food‘ maize. These biotechnologies of visibility are dependent on the use of UV lights,
without which, as can be seen in images A and B, both red and green grains remain yellow.

Figure 5.3 neatly illustrates the variety of scales at which ‗red‘ and ‗green‘ biotechnologies
are distinguished within the products of biopharming. This high-technology approach to
identity preservation is an extension of the use of more traditional approaches which ensure
―a clear distinction between transgenic plant material used for molecular farming and any
normal plant material‖ (Commandeur, Twyman, and Fischer 2003:7). As Professor B.
describes, the use of specific genetic constructs, combined with particular varieties of
plants, allows the pharmaceutical product to be visibly placed at all times:

You can use seed specific promoters, so you know your product is not going to be
anywhere else in the plant, and you can use a coloured strain of maize. I don't know
if you remember your first year genetics, you get a three to one ratio when you cross
a coloured maize with a yellow one, so you can make sure that all the seed that‘s got
your drug in it is pink. So any cross contamination of the food chain could be
immediately and easily seen.
(Professor B.; my emphasis)

In a report on containment of genetically modified crops for the UK Department of Food


and Rural Affairs, Dunwell (2005) adds the possibility of using green and purple tomato
varieties. However, these approaches to containment rely on the standardisation and
industrialisation of agriculture described by focus group participants which results in the
227
normalisation of particular varieties. They are also both culturally and geographically
specific. For example, the pictures of maize shown in Figure 5.3 rely on their distinction
from the standardised homogenous yellow variety most commonly farmed and consumed
in Europe, not the variegated varieties found in central America. Moreover, the successful
separation of the food chain from pharmaceutical is dependent on ensuring that
pharmaceutical crops can be distinguished. Although containment could be achieved
through alternative varieties, the visibility of the maize kernels in figure 5.3 is unavailable
to anyone without a UV light.

For publics, visions of biopharming are those of conventional pharmaceutical products


contained within food or agricultural hosts in which the medicines remain distinguishable
from the host plant. Edible vaccines and the blending of pharmaceutical and food
characteristics thus cause concern. The use of identity preservation approaches would seem
to reinstate the ability to visually classify pharmaceutical and food elements. ‗Identity
preservation‘ was introduced to groups in a list of potential containment approaches
presented in response to concerns such as those discussed above. In Group D (older rural
women), Hilda is particularly positive in her assessment of this approach:

Hilda: That thing there, using genes to make the crop look distinctive, now, I don't
think that's bad, because like you said, a white tomato or a red one, you do know
then, if you planted everything and they all looked the same and they weren't the
same, how would you know? I think this is what people are worried about.
(Group D, meeting 2; my emphasis)

Hilda highlights the importance of the appearance of food as a means of ascertaining its
quality. Re-establishing the transparency of products allows the reassertion of discerning
consumer behaviour. However, the visibility afforded by the superficial aesthetic that
enables consumer choice contrasts with the micro-scale of transgenes marked with DsRed
discussed by researchers. While molecular and phenotypic expression of genetically
modified identities allow pharmaceutical crops to be separated, they are not equally
accessible to consumers and producers.

Through identity preservation in the form of white tomatoes, the materials of PMPs again
become legible to consumers. However, the ability of identity preservation to distinguish
228
pharmaceutical and food products, and to assure food quality is reliant on the behaviour of
both consumers and of plants themselves. Moreover, the positive aspects of coloured
varieties which enable consumers to see contamination are questioned by Group A, who
reintroduce the pharmaceutical nature of the product to challenge the value of transparency
and visibility.

Mark: What are coloured varieties?


RM: Well, for example, if you modified a white tomato, you'd be able to see which
ones were modified
James: I don't think that would work as a good method of containment, say you
create blue maize, someone comes along and says oh my God, there's one strand of
blue maize in my crop field, then you have nationwide panic of this maize is
spreading everywhere, you know, I think because you can see it, you'd have masses...I
mean the bird flu stuff that‘s going on now, I mean one bird has it in this country this
morning, yesterday morning, and everyone's panicking and culling them
(Group A, meeting 2)

Group A problematise the social reception of identity preserved varieties. Rather than
enabling assessment of food quality, James suggests that making pharmaceutical products
visible will result in ‗nationwide panic‘. Drawing on the example of an outbreak of H5N1
avian influenza in the UK in early 2007, he suggests that this would result in ‗irrational‘
fear from other publics. His concerns reveal the extent to which traceability and identity
preservation assume particular consumer and regulatory responses. However, the
irrationality of these responses, the ‗panicking and culling‘ described in the context of bird
flu and the potential panic caused by the spread of ‗blue maize‘ make this problematic as a
means of sorting pharmaceuticals from foods.

While Hilda and James emphasise the role of identity preservation in re-asserting consumer
power, with divergent consequences, focus group discussion of identity preservation also
emphasises its uncertainty. As described in the extract from Group B below, the ability to
distinguish the products of biopharming from those of foods is reliant on trust in the
technology. They suggest that the ‗irrational‘ responses described by James may be due to
an assessment of the ability of these methods to effectively separate foods and
pharmaceuticals:

229
Evie: How do you call that word when cells change?
Sarah: Mutate
Evie: Yeah, you don‘t know what‘ll happen, even if you do coloured varieties or
maybe infertile varieties become fertile, so you don‘t know
Sarah: Well I mean there‘s cross pollination isn‘t there and all sorts
Evie: Hmm, you don‘t know, so…
Rita: It seems like the best bet, but its interesting that even that one
Jo: You can‘t guarantee it
Rita: No guarantees
(Group B, meeting 2)

The mutability of cells contributes to scepticism about establishing the visibility of


biopharmed products. The ability to separate pharmaceuticals from foods consistently is
threatened by the openness of coloured and infertile plant varieties to change.

While mechanisms of tracing and identity preservation allow the individual carriers of
transgenes to be tracked and identified, they are not equally accessible to producers and
consumers. Rather than enabling consumer distinctions between foods, identity
preservation with UV light separates the visibility of pharmaceutical products from that of
foods. As such it does little to enable consumer authority, but rather reinforces the position
of producers in a similar way to the post-marketing monitoring of pharmaceutical products.
The approach of focus group participants to identity preservation emphasises the
difficulties of reasserting the ability to discern between foods and pharmaceuticals.
Consequently, the need to effectively and reliably sort foods from medicines remains. As
suggested in James‘ comments, these are exacerbated by the co-existence of food and
pharmaceutical production within the same space, in this case that of the field.

5.7 Summary and Conclusions

This chapter has discussed the ways in which similarity and difference are established and
distributed between foods, medicines and plant made pharmaceuticals. It contributes to
understanding the second way in which the promise of biopharming is placed as the
rhetoric of the previous chapter becomes embodied in plant made pharmaceutical products,

230
as the relationship with foods and medicines becomes concrete, and as publics and
regulators encounter the technology. The key points are summarised in Figure 5.4. below.

Figure 5.4 Summary of Chapter Five


Pharmaceuticals Foods

Experts emphasise the equivalence of plant Foods underpin the promise of biopharming –
made pharmaceuticals to existing they are pure and safe, and require little
biopharmaceuticals. processing.

This is made problematic by the material The ‗natural‘ characteristics of foods


differences between plant and mammalian guarantee safety
Experts

proteins and the emphasis placed on them by


regulators and industry. However, as the example of edible vaccines
highlights, processing can not easily be
Natural proteins represent the base for entirely removed.
comparison of pharmaceutical products. They
are the metric against which pharmaceutical Identity preservation approaches to separating
quality is judged. food and pharmaceuticals require high
technology, and are culturally and
geographically situated

Focus group discussion approaches foods as


Focus group discussion draws on information consumers, who draw on a range of
provided by medical institutions and actors information in making choices

Biopharming‘s products are initially imagined, For publics the characteristics of foods are the
and are represented, through the pills and primary means of classifying them in relation
tablets of conventional pharmaceuticals to their expected effects. Secondary
information is less trusted.
Naturalness features as an important
Publics

characteristic of herbal medicines, but not as ‗Natural‘ and ‗real‘ feature as normative
an indicator of pharmaceutical quality, where comments on food quality. Industrial food
it is replaced by testing. production and processing represents a
challenge to this, as does GM.

When the example of edible vaccines is


introduced, concern emerges about the
conflation of pharmaceutical and food
qualities, and the inability to effectively
distinguish between drugs and foods.

Throughout this chapter, I have described how the meanings attached to the materials of
food, pharmaceuticals and biopharming change as they move from expert to public spheres,
from biomedicine to food and vice versa. I considered how the material characteristics of
foods and medicines contribute to classifying them in relation to normative understandings
231
of food and pharmaceuticals and to associated expectations of healthiness, safety or
efficacy. Starting with publics, I considered how expert understandings of the essential and
‗natural‘ characteristics of food and pharmaceuticals contribute to the elaboration of
particular forms of PMP. However, I introduced how expectations of safety or efficacy
associated with foods or plant-glycosylated proteins are challenged by the natures of the
products themselves and their encounters with networks of mammalian and human
biologies and consumption practices.

In the previous chapter, wider publics were enrolled as active participants in a ‗community
of promise‘ in the pursuit of pharmaceutical visions. Here I re-described the role of publics
in the research, reintroducing their role as consumers of pharmaceuticals and foods. For
these publics, expectations about the effects of pharmaceuticals and foods are established
through the information available. However, while the appearance of foods presents
information about their qualities and allows participants to act as informed, discerning
consumers, discussion of pharmaceuticals‘ qualities relies on the information provided by
trusted expert bodies.

However, when food and pharmaceutical products are brought together, knowledges of
food and farming once again become relevant. In the case of edible vaccines, the
appearance of foods and medicines is conflated. For researchers, consumer concern is
accompanied by problematic patient bodies which are unable to discriminate between food
and pharmaceuticals, leading to problems of tolerisation. For publics, the ability to
distinguish between foodstuffs and the pills or tablets of conventional pharmaceuticals
introduces new concerns about the mixing and indistinguishable blending of foods and
drugs.

In the subsequent and final empirical chapter, I consider in more detail the relationship
between food and pharmaceutical consumption and production. In particular, I extend the
discussion introduced in section 5.6 to consider how effective divisions are introduced and
maintained between pharmaceutical and food applications in the face of biopharming‘s
challenge to these categories. I focus on a final role of ‗placing‘, that which introduces the
physical locations of foods, pharmaceuticals and biopharming.

232
CHAPTER 6: PLACING PHARMACEUTICAL
PRODUCTION

I have explored how biopharming is ‗placed‘ by experts and publics through the association
of its narratives and products with those of pharmaceutical production. For experts and
publics the promise of the technology derives from imaginaries of agricultural production
and of global health. The characteristics of food crops, particularly maize, are mobilised in
the pursuit of this vision, supporting both the imagined scale of production, and the
inherent safety of plant derived biopharmaceuticals. However, the combination of food and
pharmaceutical materialities introduces new relationships between the imagined spaces of
biopharming and the material geographies of existing production regimes. I bring together
perspectives from the geographies of science with those of food to consider how local
geographies of production establish not only the quality of food for consumers but also
allow the circulation of standardised pharmaceutical products. This is accomplished
through the establishment of closed, contained spaces which contrast with those of the open
fields of biopharming‘s agricultural imaginary.

I examine the role of place in public discussion of pharmaceuticals and foods and highlight
how focus group participants emphasise the role of place in assuring quality. I suggest that
this incorporates places of both consumption and production and consider the role of these
places in focus group discussion of pharmaceuticals. I then study how expert
understandings of the features of agricultural and pharmaceutical place shape the
development of biopharming. I examine how expert and public framings of biopharming‘s
places are reconciled through an emphasis on containment. Finally I discuss the
consequences of the placing of biopharming for the realisation of its promise.
6.1 The Landscape of Foods and Pharmaceuticals in Public

Place is increasingly integral to understandings of ‗quality‘ in food production (Sonnino


and Marsden 2006), yet its role in discussion of pharmaceuticals has been less widely
investigated. Franklin (2006) and Stephens and colleagues (2008) provide one approach to
the micro-geographies of pharmaceutical innovation, whereas others have focused on the
globalisation of clinical trials and of ‗biocapital‘ (Rajan 2006; Petryna 2005). However,
there has been little discussion of the role of place in pharmaceutical consumption. Here I
examine the relationship between the places of food and those of drugs for focus group
participants.

6.1.1 The Places of Food

The relationship between place and quality was reflected in the props proffered to groups.
One of the most discussed was a Ginster‘s Cornish Pasty whose packaging epitomises
efforts to tap into the valorisation of ‗local‘ food production through the use of
‗geographically charged‘ imagery and language (Feagan 2007). Focus group discussion of
the pasty considered this imagery in detail, relating it to their own experiences of the places
of food. These exchanges define the character of food through the local and regional
geographies of its production. The places of production discussed by group participants are
dominated by those such as bakeries at which memorable meals have been bought, as in
Group A (young urban male):

Will: Is that the Cornish flag? That‘s unusual


Mark: I suppose they're trying to steal a bit of the great tradition of Cornwall
[…]
RM: So would you be more likely to get a pasty because it said it was Cornish?
Euan: I think it would be a slightly appealing aspect to it: I've been to Cornwall and
had some really nice pasties there, from some sort of shops where they were clearly
homemade, so to me as a personal thing would appeal
(Group A, meeting 1)

This exchange is prompted by Will‘s observation that the Ginster‘s packet displayed a
Cornish flag in what Mark suggests is a marketing attempt to ‗steal‘ the tradition of Cornish

234
pasty production. However, placing the pasties is complicated, and involves a combination
of historical and material characteristics. Similar discussions were also seen in Groups B
and C, where associations of place and production were strongly felt to contribute to the
appeal of the pasties. Will continues by introducing the importance of observing
production:

Will: There's a brilliant pasty shop on the north Cornish coast, well known, but I
can't remember the name of it... St Ive's. There's a brilliant pasty shop there, where
you can see it all being cooked.
(Group A, meeting 1)

Similarly, Sarah in Group B (young urban female) describes how pasties in her local shop
in North London

…would be more processed than if you actually are in Cornwall and you go to a
Cornish bakery and see pastry being made and you see it.
(Group B, meeting 1)

Both Will and Sarah suggest that the ability to witness production contributes to assuring
the quality of the product. They emphasise the importance of visibility and openness of
production. Group D (older rural women), reject the Ginster‘s pasty, arguing that the only
pasties they would eat were those they had made themselves. For Vera, this is also driven
by the visibility of production. Again, the ability to witness production is a key feature of
the places of food production and contributes to understandings of food quality:

Vera: It comes on television and shows them making them [pasties], and that puts me
off when you see all this meat and, oh no, it‘s not right
(Group D, meeting 1)

While Groups A and B contrast the Ginster‘s pasty with visible and accessible food
production, for Vera the television mediated visibility of pasty production contributes to her
rejection. As the short quote from Vera suggests, not all spaces of food are the open,
observable ones described by Groups A and B. Instead, media depictions reveal previously
closed and hidden places of production. The value accorded to open and traceable food

235
production can be seen by the negative discussion of enclosed and hidden places. These
were particularly dominated by examples of the factory and the battery farm.

The relationship between open and closed places of food production is expanded in Group
F, the young rural mixed group. Participants in Groups A, B and D above describe the
importance of the visual features of production space. Group F problematise this by
describing the presentational work that goes into establishing links between place and
quality. In the following extract, open spaces such as fields and the countryside are
contrasted with small pens and ‗inside‘ farming. As for Vera, knowledge of the closed
places of agriculture is mediated through a TV programme, rather than being directly
observed as are the Cornish pasty shops of groups A and B. In the absence of access to
production sites, the places in which food is encountered, purchased and consumed act as
‗spatial proxies‘ for food quality. Open spaces are linked with farm shops and butchers,
while supermarket food is ‗shocking‘ and conceals contained and constrained farming
practices.:

Sam: Well where are you going to see the ones, see the pigs that are not in fields
Katie: But I don't think they are
Rebecca: There was a programme wasn't there
Sam: I think we're all just, you know we live in the country and everything's rural and
lovely...
Rebecca: There was a programme...
Sam: The majority of pigs in England don't live in fields
Max: They're in little pens aren't they
Sam: Yeah, and they're inside aren't they, we see them in Bury don't we, because it‘s
very rare, and it...
Josh: The ones in the fields are for breeding aren't they
Sam: And also for farm shops and butchers, decent butchers, not what you buy from
Sainsbury's, that'll be shocking
(Group F, meeting 1)

These extracts illustrate the importance of openness to food quality, and the association of
closed production with particular forms of agriculture. In Group C‘s (older urban mixed)
discussion of Cornish pasties, the relationship between open and hidden places of

236
production is expanded as Ian describes his experience of working in a factory producing
food:

Ian: There's a certain danger I think to having foods like that [pasties], even though I
eat them myself, because, I've worked in a factory before, where pies and that lot
have been made, and sometimes people just aren't as caring as they perhaps should
be, and that is my own experience
(Group C, meeting 1)

Ian‘s discussion of pasties brings closed production together with that of the aesthetics
discussed in Chapter Five. The fact that the product is ‗all wrapped‘ and his own experience
of working in a food factory contribute to his scepticism about the pasty‘s quality.

Throughout these extracts, discussion is bounded by groups‘ own placed experiences of


food production and consumption. Group F‘s discussion is situated in relation to their
environment as country-dwellers, where everything is ‗rural and lovely‘. Groups D and F,
which are more sceptical about the ability to judge production, are rural, whereas Groups A
and B, both urban, seem more convinced of the power of witnessing production but contrast
it with their daily routines. For Group C Ian‘s experiences attest to factory production and
make it visible to the group.

The places of food production, processing and consumption are a central part of the
assessment of food quality by focus group participants. The discussion in the extracts
presented above echoes the importance attributed to location in descriptions of
contemporary agro-food geographies (e.g. Murdoch 2005). The importance of
pharmaceutical place to publics has been much less examined, and it is to this that I now
turn.

6.1.2 Pharmaceutical Places

Public discussion of the geographies of pharmaceuticals is generally absent from social


science research. Focus group discussion offers some explanation for this. For Will, a
participant in Group A, it is the control of potential side effects through pharmaceutical
testing and regulation that allows the provenance of drugs to be obscured. The

237
standardisation and classification of the pharmaceutical product ‗dis-places‘ the production
process. As such, it allows medicines to circulate in what Murdoch (2005) describes as
‗topographies‘ which ensure ―predictable outcomes in diverse local circumstances‖ (2005:
168):

Will: I generally work on the assumption that medicines off the shelf, wherever they
come from, are far more regulated than food, and to get there they have to go
through quite a lot of stringent testing, so if I've got a cracking headache or a
hangover, really I don't care where it comes from, I'll just grab whatever headache
tablets I can, because I feel that it’s been tested correctly
(Group A, meeting 1; my emphasis)

Testing and regulation, the processes of ensuring equivalence between pharmaceutical


products, distinguish them from foods. Trust in regimes of testing and regulation stand in
for the ability to individually assess the qualities of pharmaceuticals. As seen in this extract,
they also allow the circumstances of pharmaceutical production to be obscured. This
contrasts with the same participants discussion of the places of Cornish pasty production.
As Group F continues in the extract below, pharmaceutical origins are not something that is
generally considered:

RM: What about this thing of who makes things and where it comes from, like with
food. Does that matter for medicines?
Rebecca: I don't really think about it
Max: Because they have to go through such rigorous approval tests and everything
else
Katie: I wouldn't have a clue
Sam: We could not know
Max: Yeah, we could all be very naïve
Katie: I don't know, don't you think the fact that it‘s got Boots on it, makes you it
should be alright, but because it‘s a brand name that we know, it‘s like Tesco's or
Sainsbury's or something I'd think it'll be fine, if it was Morrison I might think I'm not
going to touch it!
Max: I'll take that on the food argument, but not on the drugs
[pause]
Max: Most of us haven't even thought of it have we?
(Group F, meeting 1; my emphasis)

238
For Max, the clinical trials and testing procedure for pharmaceuticals ensures their quality
and negates the need for assessment of production places. The contrast between the role of
place in foods and medicines is explicitly opened up by Katie and Max. When Katie
attempts to extend her discussion of the role of different places in her assessment of food
quality to that of pharmaceuticals, Max explicitly rules out the comparison. In these
extracts, pharmaceutical quality control is central to the separation of medicines from their
place of production. The absence of discussion of these places leads to increased faith in
and reliance on this quality control as the guarantor of safety and quality.

For Group E (older mixed rural) there was also little awareness of pharmaceutical
production, although rather than representing a lack of knowledge this is due to the
availability of choice and the mediation of this choice by doctors:

Helen: I don’t think you have the choice do you, so we haven’t even begun to think
about that one have we? We‘re prescribed something and you don‘t get asked do you
want it from here or there do you
Joan: If you‘ve a regular medication…
Jane: Most drugs companies are foreign owned, and anything that‘s produced here is
under licence so there isn‘t even the economic interest for this country - a lot of stuff
comes out of Ireland.
(Group E, meeting 1; my emphasis)

Helen suggests that the absence of pharmaceutical places from discussion is dictated by the
lack of choice in pharmaceutical consumption. For focus group participants the places of
food vouchsafe its quality and its safety. In contrast, for conventional pharmaceuticals
―we‘re prescribed something‖. The places of pharmaceutical production are occluded by
those of prescription. Instead, when Jane introduces specific places of pharmaceutical
production they relate to concerns of political economy, rather than to individual choice and
to the individual facilities that characterise discussion of food.

The extracts presented here emphasise that the places of pharmaceutical production do not
feature within the discourses of everyday life. In contrast, both practices and places of
production and those of consumption feature heavily in discussion of foods. Two types of
spaces of food production are described, those which are open to the consumer and those

239
which are closed. The visibility of production contributes to trustworthiness, whereas
hidden spaces of production are perceived much more sceptically.

6.2 Biopharming’s Public and Expert Places

Placing biopharming among the ‗locations‘ of food and medicines is performed in


contrasting ways by publics and experts. The absence of pharmaceutical place from focus
group discussion is continued as participants consider biopharming, and for publics the
most important places of biopharming are those which allow it to be separated from the
agricultural and food places described above.

6.2.1 Publics: The Pharmaceutical Places of Biopharming

Pharmaceutical geographies are not prominent in focus group discussion. When pressed to
consider similar the places of medicines as they do those of food, focus group participants
emphasise the importance of regulation, standardisation and prescription. This is continued
in initial discussion of the places of biopharming, as introduced in discussion from Group
D:

Sofia: You're going to be very very busy Richard, you're going to have to go all round
and get groups like this and then it'll start thinking, start them all thinking you see,
because honestly, I've never even thought about, you just take your medication don't
you, until you read what the side effects is and then you're frightened to death
Molly: You don't know where they come from really
Elsie: But you don't think how it‘s made or
Sofia: No I think, it‘s made us all think, but you'll have to go round…
(Group D; meeting 2)

As this group emphasise, the production origins of pharmaceuticals do not form part of
everyday discussion, and only arise when participants are prompted to discuss
biopharming. Instead, as Molly and Elsie describe, they do not know and do not think about
where pharmaceuticals come from. This ignorance of the origins of pharmaceutical
products is voluntary, in that it is not thought about, rather than hidden in the same way as
factory farming. This is a form of ‗functional cohabitation‘ (Michael 1996) with
240
pharmaceutical science, in which further information beyond that about side-effects is
avoided.

When individual spaces related to pharmaceutical production are discussed in the context of
biopharming, it is due to the breakdown of the seamless movement from research to
pharmacy shelf, when the mechanics of testing and standardisation fail. The most
prominent example of this in focus group discussion is the failed clinical trial by German
biotechnology company TeGenero of monoclonal antibody TGN1412 at London‘s
Northwick Park hospital in 2006, which endangered the lives of participants and made
national news headlines. As do food scares, this trial opens the ‗black box‘ of production to
reveal the ‗hybrid collectives‘ (Goodman 1999) of people and places which lie behind the
foods and medicine displayed as prompts. A second extract from Group D emphasises the
role the trial plays in placing pharmaceuticals:

Hilda: Sometimes you can't help going back to - you know that trial that they did, was
it last year, when they had that special trial? Nothing to do with this mind you. They
had those people and they nearly died, in London
Molly: Oh yes
Hilda: And you think, well my mind‘s gone back onto those sort of things you know,
and they...
Jean: What was that for, was it a common cold?
Hilda: I can't remember now what it was for, but they took these people in, and they
got money for doing it you know, and, well I think one of them will never work again.
But I think really, seeing that in the paper, if you don't know anything about these
things you wouldn't bother would you, you wouldn't, but now we're more interested,
so you know, you feel more interested in it really. You're aware of it.
(Group D, meeting 2)

For Hilda, the TeGenero trial provides a way into talking about the potential risks of
producing pharmaceuticals in plants. It places and exemplifies a process with which the
group was otherwise unfamiliar. Generally, testing and clinical trials allow pharmaceuticals
to circulate as standardised and placeless products within ‗topographical‘ (Murdoch 2005)
regimes of production and consumption. In the case of the Northwick Park trials the
topologies of pharmaceutical innovation were laid bare, linking German biotechnology
company TeGenero to clinical trials facilities in North London. Relations between
241
TeGenero, trials company Parexel, NHS hospitals and regulatory agencies were opened up
to media and public scrutiny, and the economic, geographical and social distribution of the
clinical trials process was highlighted (see for example Goodyear 2006).

While Group D describes biopharming in terms of the TeGenero trial and existing
biopharmaceutical production, Group A introduces agricultural places to contrast
biopharming with these existing models. In the extract below, the risk of contamination of
food and the environment divides biopharming and existing forms of pharmaceutical
production. The novel characteristics of biopharming in maize, its mobility and scale,
present novel challenges for controlling pharmaceutical facilities:

Rob: Pigs are easier to control aren't they? You can regulate the number of them.
Maize can spread. If you have a field of maize plants, I'd imagine they'd be harder to
contain
James: Maize doesn't move, it doesn't need feeding, whereas pigs do
Rob: Yeah, but they like reproduce don't they, pollination or something, I don't know
how they, I don't know how maize actually does reproduce, but if it did, you know. As
far as I remember it only takes a bee to land in a flower and move to another one
Will: So you're worried about the outbreak of maize are you?
Rob: No, but there's...if you're worried that you're interfering with nature, you might
create some dodgy plant that ends up spreading and, I don't know, taking up all the
soil space that other farmers might have been using to grow other crops for more
food
(Group A, meeting 2)

The openness, but also restricted availability, of field space is used to support a contrast
between biopharming in maize and the one example of existing production provided in the
booklet, that of gastric lipase, currently extracted from pigs. The ability to control the
movement of pigs is contrasted with the recalcitrant and unruly nature of maize. Discussion
is framed by an understanding of the characteristics of arable production and arable space,
the field, as open and uncontrollable.

The ability of maize to spread and appropriate the space of food production forms the core
of Rob‘s concern about biopharming production. Unlike existing pharmaceutical
production, maize plants are less amenable to control, and exist in close spatial relation

242
with existing agriculture. The openness of agricultural, and particularly arable, spaces, in
which insects and plant seeds are able to move freely, constitutes them as vulnerable spaces
in which food production can be usurped by pharmaceutical production.

Focus group participants struggle to relate biopharming to the places of existing


pharmaceutical production, primarily because ‗normal‘ pharmaceutical innovation is
hidden and placeless. It is only when the standardised and homogenised networks of drug
manufacture break down that they are open for inspection by members of the public. In
contrast, as focus group talk develops, Rob introduces a detailed discussion revolving
around the challenges biopharming presents to the places of food and agriculture.

6.2.2 Publics: Biopharming and Food Spaces

The dominant places of biopharming in focus group conversation are those of agriculture.
The relationship between place and food safety and quality is reflected in concern about the
separation of the places of food agriculture from the risky natures of biopharming. This is
introduced in the extract below from Group B:

Rita: I think the question of contamination is … the biggest consideration there is the
question of degree. Yeah, quantity, amounts, and contamination. It is a problem that
will build up over time, and it, the problem starts small, the situation starts small, and
then it grows, I think that is one of the big considerations
RM: So…
Rita: It might be small quantities at the beginning, and then the quantities keep
increasing over time. And then that is one of, one of the real dangers that will creep
in. You know, as I see it, so it might not be something that manifests at the beginning,
it might be something that takes a little while to develop
Jo: It does take me back to what we were talking about last week about how much
actually people do think about what does go into food production and, I mean I guess
unless you‘re going to grow your own food and you know, produce your own oils and
everything, you can‘t really be in control of what, you do have to trust, or you have to
not think about it too much
(Group B, meeting 2)

Group B makes explicit the relationship between concerns about development of


biopharming and those introduced in their previous meeting. As Rita describes, the main
243
concern about biopharming is the contamination of the food supply, and its augmentation
over time. The risk from pharmaceuticals is seen in terms of a gradual leakage or pollution
which builds up to hazardous levels, rather than a one-off event.

Discussion of biopharming in this group relates to the difficulty of assuring food quality
and safety, and to the ability to control agricultural production. Separation between the
group as consumers and the production of their food contributes to mistrust. Trust in
production and wilful ignorance are described by Jo as the only alternative to taking
complete control of one‘s own food supply. Ignorance of food production represents an
attempt to cohabit, but unlike in discussion of pharmaceuticals, it is as a coping strategy, as
a deliberate choice rather than an accepted norm.

While agricultural places are normally associated with food quality, the driving concern for
groups in their discussions of biopharming‘s places is its potential threat to food safety. The
potential for pharmaceutical contamination of the food supply, requires the separation of
food and pharmaceutical products. In group discussions, the preferred approach to achieve
this is the separation of agricultural and pharmaceutical space. These challenges to the
development of biopharming temper appreciations of its potential, as in Group D:

Sofia: I do think it‘s not just straightforward like we think, planting things, it‘s like
the, like they say, the pollen, they‘d have to be…and things that are mixing together
with other plants you see, growing, so I don‘t think it‘s, there‘ll be a lot they have to
test, it‘ll be a long while before its safe
Hilda: It won‘t be in our time will it
(Group D; meeting 2)

The need to establish the safety of biopharming in relation to food production shifts the
promise of the technology into a more distant future. The group presents testing as a means
through which relations between the places of biopharming and those of other plants can be
stabilised. As for pharmaceuticals, the testing and standardisation of biopharming would
remove concerns about its places. However, for other groups the complexity of agricultural
space raises problems with testing, as in the extract below from Group A:

Rob: I'd like to know what would happen to the animals that eat the maize, like field
mice and things like that, and the insects that feed on them, like how it would impact
244
on the food web and sort of things like that, I don't know maybe that...before it was
er, used widely
Mark: But that's part of the problem, it‘s things like that that are really hard to find
out, isn't it, because it‘s so complex
(Group A)

The complex associations of agricultural space with a diverse array of actors challenges the
ability to place biopharming‘s relations with this space. This discussion is developed in the
extract below from Group C, which arises when the group is presented with a quote from a
Greenpeace report on molecular farming. Group C is the only group to display sympathy
for the position put forward in this statement, and indeed a number of groups discuss the
organisation negatively. However, here the group discusses field trials as a possible means
of ensuring the safety of biopharming, and consequently focuses on molecular farming‘s
relationship with agricultural biotechnology.

RM: Ok, what about this one, ―These plants could cause irreversible harm to the
environment‖, and that‘s Greenpeace
Ian: I‘m kind of in agreement with that
Catherine: See that‘s in a sense what I‘m saying, devastating consequences, that‘s
exactly what I‘m saying, see
Peter: They‘re not even open to testing, there‘s not even a sense they‘re saying, well
if they‘re tested we might consider it, they‘re just…
Ian: I mean I would be on the same kind of thing, because I would be really, really
err on the side of caution. It is…
Catherine: We don‘t know what we‘re doing to the environment, I‘m sorry Peter
Peter: Then we‘ll test it
Catherine: Well then, once we test it, that‘s it, there‘s no going back, we‘ve crossed
the Rubicon
(Group C)

Faced with Peter‘s insistence on testing, Catherine challenges the ability of testing and of
field trials to divide experimental agricultural and food agricultural space. She suggests that
the lack of knowledge about the environmental consequences of biopharming can not,
paradoxically, be answered without these very consequences being risked, due to the place
of the experiment. In Catherine‘s terms, testing, not deliberate release, represents ‗crossing
of the Rubicon‘, or the ‗irruption into social space‘ described by Babusiaux et al. (2003).
245
Ensuring the quality of food is predicated on the complete removal of biopharming
experimentation as well as production, from agricultural space. The testing that enables
pharmaceuticals to circulate in placeless topographies is less persuasive and less feasible in
an agricultural system.

I have so far focused entirely on public discussion of the relationship between biopharming
and the places of food and medicine. I have suggested that for publics places are central to
assuring the quality of food. However, there is no parallel discourse around
pharmaceuticals as regimes of testing and standardisation contribute to obscuring
pharmaceutical place. Concordant with this general emphasis, the places of biopharming
are described in relation to those of agriculture. The importance of these places to
biopharming challenges the ability of PMPs to become displaced in the same way as
existing pharmaceutical production.

6.2.3 Experts: Biopharming and Agricultural Space

The food and agricultural characteristics of biopharming are central to its promise. Not only
does agricultural production form the basis of the potential for large scale, low tech
production, but for experts similarities with foods render PMPs safer than their bacterial or
mammalian counterparts. However, PMPs raise significant challenges to the integrity of
networks of food and pharmaceutical safety and quality. By taking on the characteristics of
agricultural production PMPs become self-reproducing and stable. In contrast, Chinese
hamster ovary cells or E.coli struggle to survive outside the bioreactor. The agricultural
nature of PMPs both constitutes and threatens their promise.

In their contribution to Molecular Farming (2004), Twyman and Commandeur highlight


two sets of risks posed by pharmaceutical production in an agricultural setting, those of
transgene spread and of unintended exposure. As they point out, these are not easily
separated, as the former can lead to the latter:

―because the naturalization of transgenic plants outside the intended production site
results in the wider exposure of non-target organisms. While these risks apply to all
field-grown transgenic crops regardless of their use, those used for molecular

246
farming deserve special attention because of the pharmacological or toxic properties
of many of the recombinant proteins they produce‖
(Commandeur and Twyman 2004:252)

This quote highlights the complexity of farming pharmaceutical molecules and the
intersection between the products described in the previous chapter and the places
described here. While pharmaceutical crops raise the same problems as other transgenic
varieties, these are exacerbated by pharmaceutical genes. The risks operate at a number of
levels: there is not only the pharmacological and toxicological threat to the systems of those
exposed as ‗non-target organisms‘, but also the size at which production takes place, that of
the field. In the same edited collection, Brandle describes how: ―it is critical to keep crops
producing such materials out of the food and feed chain, and the environment‖ (Brandle
2004:71).

For PPC experts, encounters between biopharming and agriculture are framed by past
experiences of the biopharming industry, particularly that of Prodigene22. In the ‗Prodigene
fiasco‘ (Schiermeyer, Dorfmuller, and Schinkel 2004), failure to sufficiently clear fields of
biopharmed maize plants led to the contamination of subsequent soya crops. As a
consequence, US biopharming firm Prodigene was heavily fined and eventually went out of
business, while the US regulatory environment was significantly tightened (Schiermeyer et
al. 2004).

The Prodigene case is particularly apposite to discussion of PPC. Prodigene was a pioneer
in developing maize production systems, and as such many of the concerns for Prodigene
are echoed for the PPC. As Ma and PPC colleagues describe in a 2003 paper:

―Recently publicized incidents in which genetically enhanced crops have been


inadvertently mixed with those destined for human consumption have highlighted the
need for mechanisms to ensure the segregation of plants that express
pharmaceuticals‖
(Ma et al. 2003:85)

The Prodigene case highlighted the importance of distinguishing between molecular


farming and other agricultural practices. In summer 2008, the EFSA published a ‗Draft
22
see Chapter One, section 1.6.1 for a fuller discussion
247
Opinion‘ on the risk assessment of non-food and feed agriculture which directly addresses
biopharmed crops. Although no pharmaceutical crops have yet been authorised for
commercial release, up to this point trials have been dealt with on a similar ‗case by case‘
basis to existing genetically modified varieties. The document suggests that for the EFSA:

―A major issue to be addressed during the risk assessment is that GM plants and the
derived material used for the production of industrial or biologically active/medicinal
products may potentially enter the food or feed supply chain (via admixture or gene
flow) or may be accidentally consumed from the field”
(EFSA 2008:10; my emphasis)

The risk highlighted by the EFSA is that of contamination and accidental consumption due
to the inability to distinguish between pharmaceutical and food products. As described in
the previous chapter‘s discussion of edible vaccines, for publics, the similarity of
biopharming‘s products to foods represents a potential challenge to the technology‘s
promise. Here, the places in which biopharming occurs become central to the separation of
agricultural production of pharmaceuticals from that of foods.

The EFSA‘s division of the risks posed by biopharming into contamination and accidental
consumption echoes that proposed in papers by PPC researchers, notably Commandeur and
Twyman (2004). For the EFSA (2008), the assessment of risk involves a molecular
characterisation of the ‗transformation event‘. As with first generation genetically modified
crops for food and feed, dealt with under guidelines 1829/2003/EU, this establishes the
‗event‘ of the GM plant as its focus (Lezaun 2006). The transformation event becomes the
focus of a topology of traceability, in which the act of genetic modification can be
followed. However, for biopharming researchers, not all the results of transformation
events are of equivalent interest, as described by Professor A.:

You don't want to risk contaminating the food chain with something, and you can see
that that would be a problem with something that was very pharmacologically active
(Professor A.)

For both regulators and researchers, the potential contamination of foods with
pharmaceuticals is an important factor in the development of biopharming and in the
elaboration of a place for the technology. However, in this quotation from Professor A. a
248
distinction is opened between the products of biopharming. The separation between
antibodies such as that produced by the PPC and more pharmacologically active antigens is
frequently introduced in interviews. While the former are adjudged benign on the basis of a
tradition of consumption, the latter require significantly greater vigilance, as described by
Professor C.:

It will depend very much on the product you see. If it‘s an antigen, those are
potentially quite toxic and quite nasty, and so that will need to be probably always
grown under containment, or very closely defined confinement, you know in really
way out places. Antibodies are much potentially much less of a problem, because you
know, when you drink milk you have lots of antibodies, and so on.…In some cases,
some antigens will probably never be fit to go outside and will always have to be
done in very, very contained, highly contained conditions, but some others may, you
know, may not be a problem
(Professor C.; my emphasis)

Professor C. here draws on arguments which are familiar from the previous chapters. The
safety of biopharming‘s products is attested by their biological familiarity and narrative
continuity with foods. However, in this extract, the researcher introduces a division
between the places of biopharming, between contained and ‗outside‘ production. The
places of biopharming are co-produced with its products as antigens such as those produced
for vaccines are singled out as requiring separation from food through containment. In
contrast, antibodies such as that of the PPC‘s HIV microbicide are benign, familiar and
unthreatening. They are ‗fit to go outside‘. However, neither Professor C. nor Professor A.
offer suggestions as to how decisions would be made on what would be a sufficiently
pharmacologically active product to warrant separation from food production.

The expert discussion presented here suggests that the separation of food from
pharmaceutical products is one spur to the placing of biopharming. The nuancing of
different pharmaceutical forms suggests that such places may take diverse forms, and that it
is difficult to imagine a singular place for the technology. However, for experts the need to
separate biopharmaceutical and conventional agriculture is complemented by that to resolve
the relationship between agriculture and biopharmaceutical space.

249
6.2.4 Experts: Spaces of Pharmaceutical Concern

Biopharmaceutical production in plants presents a novel challenge to agricultural space and


to the EU regulations which govern this space. However, perhaps more importantly,
biopharming prompts new relations with the locations of pharmaceutical production.
Equivalence with pharmaceutical quality regimes is central to the biomedical promise of
biopharming. This quality is a product of the places of pharmaceutical production, as they
are described and circumscribed by pharmaceutical regulations. In the EU, the essential
characteristics of pharmaceutical quality are described by good manufacturing practice
(GMP) guidelines, laid out in Directive 2003/94/EC and enacted into UK law by the
‗Medicines for Human Use Regulations‘ of 2005. The description of good manufacturing in
the regulations is laid out in the EU‘s ‗Eudralex‘ guidelines. In the Directive, GMP is
defined as:

―the part of quality assurance which ensures that products are consistently produced
and controlled in accordance with the quality standards appropriate to their intended
use‖
(EC 2003:2)

Achieving GMP is a combination of control over both personnel and premises, rendered
traceable through documentation and regular inspection. It involves the removal of dirt,
pollution and other matter ‗out of place‘ (Douglas 2002) in the pursuit of pharmaceutical
purity (Franklin 2006). In particular, section 2 of Article 8 of 2003/94/EC describes how:

―Premises and manufacturing equipment shall be laid out, designed and operated in
such a way as to minimise the risk of error and to permit effective cleaning and
maintenance in order to avoid contamination, cross contamination and, in general,
any adverse effect on the quality of the product‖ (2003/94/EC).

It is the problem of applying these guidelines to agricultural production that forms the heart
of the concerns described above. As Ma, Barros et al. (2005) point out, GMP was designed
for the production of chemical pharmaceuticals, and later modified for biopharmaceutical
production. Its key feature is the establishment of closed, controlled systems defined from
start to finish. They represent an idealised and highly developed version of the molecular

250
biology laboratory space described in Chapter Two, aiming factory-like at the production of
purified, uniform, mass-produced substances (Knorr-Cetina 1999).

While GMP includes a range of protocols and procedures, work by Stephens et al. (2008)
and Franklin (2006) points to the importance of the architecture and geographies of GMP
facilities. A GMP facility has recently opened at a Pharma-Planta partner institution,
Fraunhofer IME Aachen, and is shown in Figure 6.1.

Figure 6.1 The GMP Facility, Fraunhofer IME, Aachen

The facility enables PPC material to be processed at the same location from plant to
product. The form of this building, and of standardised GMP facilities in general, gives an
insight into the role of place in pharmaceutical production and in biopharming in particular.
The geography of the Fraunhofer site separates the closed site of the GMP facility and that
of the initial plant processing facilities – they are linked by the corridor on the right in the
picture. The GMP facility is described in the following terms on the Fraunhofer IME
website :

251
―2 independent multi-purpose GMP production lines for the production of
recombinant biopharmaceutical API [active pharmaceutical ingredients] are
installed in the facility…
…RO [reverse osmosis]-water, clean steam and WFI [water for injection] generators
are located in the basement and distributed to the clean room areas. Two central CIP
[Clean-in-Place] stations, also located in the basement, distribute CIP solutions to
all rooms in which CIP-processes are to be carried out. A thermal inactivation and a
cooling tank for hot waste water feed waste liquids to the central neutralization
tank…
…The general room layout has been approved by the local authorities who have also
inspected the facility in January 2006 during the installation of the clean room
walls‖.23

The description highlights the role of the building‘s spaces in establishing GMP control.
Rooms are separated from one another and are dedicated for specific purposes, while
everything that flows through the rooms is sterilised. Micro-monitoring and surveillance of
these spaces is complemented by verification and inspection by local authorities, who link
the facility to regulations issued at EU level. The visibility of contamination starts at 0.5μm
and terminates at the supra-national level. The IME facility is available for generic
recombinant pharmaceutical production, not specifically plants. As biopharmaceuticals
move into pharmaceutical facilities, both become standardised and the products are stripped
of their original circumstances of production. As Franklin (2006) describes in discussing
similar facilities in embryonic stem cells research, GMP quality assurance systems act not
on biology itself, but by controlling its milieu.

For focus groups, testing and quality control separate pharmaceuticals from their place of
production. In contrast, in expert discourse, these places are central to assuring this quality.
The promise of biopharming derives from a comparison with the restricted and enclosed
conditions of CHO and E.coli production. However it is the ability to regulate and control
these systems which guarantees the quality of the pharmaceutical product. To enable this,
pharmaceuticals must again be separated from the food supply. For Professor L., the
uncontrolled spaces of agriculture, particularly organic production, are threatening to the
safe production of medicines:

23 http://www.ime.fraunhofer.de/EN/mb/production/GMP_facility.jsp
252
If my pharmaceutical maize got contaminated by Kellogg‘s cornflake maize, I would
be upset too, because that‘s also a failing of containment, and I'd be doubly upset if
an organic farmer contaminated my maize because there'd probably be all sort of
bugs coming over from his farm. It‘s a two way thing. They're worried, and I'm
worried too about my pollen getting out, but I'm worried about their pollen getting
in, because I, as a pharmaceutical I have to rigidly control my starting material,
and I don't want any contamination. And that will be part of cGMP [current Good
Manufacturing Practice]
(Professor L.)

Contamination by the ‗bugs‘ of organic farming threatens the control required for
pharmaceutical production. The geographies and natures of agriculture which constitute the
promise of biopharming are now threatening to it. The separation of pharmaceuticals from
the mess of agriculture to establish their quality is an essential part of ‗cGMP‘. For
researchers, compliance with Good Manufacturing Practice is the driving force behind this
final placing of biopharming. Professor A. expands on the challenge for biopharming in the
extract below:

You always tend to think about from the plant side, stopping an unwanted in this case
an unwanted virus getting out or the transgenic lines pollen getting out, crossing
gene flow to the environment. Of course for pharmaceutical plants, you probably
don't want to have your nice pharmaceutical proteins growing outside, because I
mean if you think you know when you harvest plants outside there’s a lot of
basically crap on them to be honest, you know birds, you know they're not going to
be very nice and sterile systems, so you probably want them to be contained anyway
to prevent unwanted fungal toxins
(Professor A; my emphasis)

Not only does biopharming risk contamination of food and the environment, but the
environment is itself threatening to biopharming. In these terms, pharmaceutical production
is not something that can take place ‗outside‘, despite visions of fields of antibody
producing corn and however pharmacologically active or inactive the product.

Expert description of agricultural production in these terms is directly related to the EU


regulatory guidelines on biopharming, which lay out the requirements for GMP, and
describe how field space contains non-pharmaceutical material such as:

253
―extraneous material such as insect, bird and animal excreta, carcasses or parts
thereof, organic fertiliser residues, and/or production personnel-shed material‖
(EMEA 2008: 9)

Consequently, the guidelines advise that

―considering the specificities inherent to plant cultivation, particular attention should


be placed on the demonstration of the robustness of the production processes‖
(EMEA 2008:7)

As Professor A. describes in the quote above, making agricultural production systems


correspond with those of pharmaceutical production is problematic. The EMEA guidelines
on biopharming separate it into first and second or ‗upstream‘ and ‗downstream‘ sections of
the production process. Agricultural space is confined to the upstream section, allowing the
adoption of a standardised second phase involving the same facilities as conventional
biopharmaceuticals. It is during the first phase that differences between plant made and
other biopharmaceuticals are smoothed and obscured, as the draft EMEA guidelines of
2006 describe:

―The first production phase is specific to transgenic technologies and includes the
cultivation, harvest and primary processing (for example screening, cleaning,
sorting, transporting and storing) of the harvested material. The second phase,
encompassing product isolation, purification, formulation, etc., is common to all
biotechnology-derived products and the general requirements are well documented in
the relevant guidelines [on GMP]‖
(EMEA 2006:7)

The ‗first production phase‘ of PMPs involves the processes that make up agricultural
production: cultivation, harvest, and post harvest, and which delineate biopharming from
existing pharmaceutical production. However, to bring biopharming in line with existing
pharmaceutical production, these distinctive features must be controlled in the same way.

The key problem for researchers is how the agricultural spaces of biopharming are related
to ‗Great Big Steel Things‘ (Professor A.) - the cGMP-certified bioreactors which
characterise conventional biopharmaceutical production facilities. The means to do this are

254
expanded in EMEA‘s discussion of molecular farming sites. It describes how these must be
defined in a number of ways:

Geographical location, with boundaries exactly defined


The quality and nature of the growth substrate (typically soil, aqueous solution, or
aqueous suspension), water supply and other raw materials (including fertilizers
and pesticides) should be defined, and specifications should be set, where
appropriate.
The prevailing meteorological conditions, with seasonality and general variability
should be documented. Extreme conditions for the locality should also be
mentioned.
Supervision of the site
Local flora and fauna
Cultivation of other genetically modified plants in the vicinity
The quality and/or good practice system in operation at the site.
(EMEA 2008: 8)

The regulatory approach to biopharming is linked to the specific physical location of


production, the practices that occur at this location and the environmental characteristics of
these sites. They involve controlling the behaviour of humans and non-humans within a
geographical location. The guidelines outline the essential characteristics of a ‗place‘ of
biopharming. As one researcher described following a meeting with the EMEA, these
characteristics are central to a regulatory concern with the reproducible quality of the
pharmaceutical product described in Chapter Five:

These guys come from normal stand-up biotech. You have your fermenter and you
can do your analyses, and you're in a contained system and you can do your cell
banking procedures and it‘s all set in stone, more or less. And they want the plant
people to mimic this as close as possible. The biggest concern was reproducibility.
We could say whatever we wanted – ―Well we have this interesting‖..."Very nice, but
what about reproducibility?" Every second sentence was this. And they mean not only
reproducibility in the downstream process but everything starting from the
cultivation, potential influence of climate, of UV radiation, of water used for

255
watering, of fertilisers used, type of soil the plant is growing in, do they use pots, do
they use no pots and so on...the whole nine yards
(Dr J.; my emphasis)

The micro-control of pharmaceutical GMP required by regulators is alien to an agricultural


production system described in terms of sun, soil and water, and as filled with animal
carcasses and excreta. The need to reconcile these two spaces, the one controlled and
sterile, the other unruly and dirty provides the impetus for a search for a controlled
biopharming space. The exact way in which such places are to be established for
biopharming is complex, as recognised in EMEA‘s first draft guidelines, which describes
the need for ‗interested parties‘ to become involved:

―The establishment of an appropriate in-house Quality system for the first phase of
the process when GMP cannot be applied is critical for ensuring consistent
production using transgenic plants. It was considered that it is difficult to provide
more specific guidance in this area at the present time.‖
(EMEA 2006: 7)

EMEA‘s own expertise is insufficient to offer specific advice on the development of a


Quality system for biopharming. In the updated regulation, this is resolved through a focus
on the alternative traditions of botanical medicine production, as the locational ‗placing‘
biopharming becomes linked to its temporal placing as an extension of plant-based
medicines. The finalised guidelines focus on herbal medicine production methods and on
the ways in which traditional plant medicines are standardised in providing guidance for
biopharming. They describe how, in the absence of ‗classical GMP‘:

―The development of the system may use as a starting point the basic principles
outlined in the HMPC [Committee on Herbal Medicinal Products] ‗Guideline on
Good Agricultural and Collection Practice (GACP) for Starting Materials of Herbal
Origin‘‖
(EMEA 2008: 6)

The relations between herbal practices and biopharmaceutical production return in


developing the place of biopharming as regulators attempt to materialise a ‗salient historical
node‘ for biopharming. In Chapter Four, Dr H. describes how the regulations governing
herbal medicines might be more applicable to biopharming production than those relating
256
to biopharmaceuticals. However, other researchers see these regulations as inadequate and
insufficiently controlled to function as a pharmaceutical system. This is also the case in the
EMEA guidelines on molecular farming, which continue:

―Confirmation of compliance with the GACP Guideline, which is aimed at a different


usage of plants, is not alone considered adequate for controlling transgenic plant-
based production.‖
(EMEA, 2008: 6)

Plant made pharmaceutical production is established as discontinuous from herbal


medicines on the basis of its process and projected use. GACP can not constitute a ‗quality
system‘ (EMEA 2006) for biotechnological medicines, as Dr K. describes:

The transgenic plant field is completely different from herbal medicines. They
[EMEA] say you work with GMOs and this is biotech and so biotech applies.
(Dr K.)

As Jasanoff (1995) describes, the process of biotechnology frames EU regulatory responses


to agricultural biotechnology and separates it from existing food production. Similarly, in
EMEA‘s approach to biopharming, the biotech process separates it from the production of
herbal medicine.

6.3 Reconciling Agricultural and Pharmaceutical Spaces

Agricultural and pharmaceutical characteristics combine to drive the framing of


biopharming in terms of containment. Both biological and physical containment methods
have been proposed. However, as I explore here, not only must pharmaceuticals be
excluded from foods, but for experts, food and agriculture must also be separated from
pharmaceuticals. As such I now focus on how physical containment approaches enable
biopharming to be placed.

6.3.1 Publics: Located Promise

Focus group discussion of the places of biopharming concentrates on the risk of


pharmaceutical contamination of the food supply and the environment. Among the props
257
presented to focus groups in the second meeting was a list of possible containment methods
shown either when discussion moved to consider the problems of separating food and
medicines or after the interval in focus group discussion. These included both biological
and physical containment methods, some of which have been discussed in previous
chapters. Of the latter, greenhouses are discussed most, as in the extract from Group D
(older rural female) below:

Sofia: You can‘t just grow tobacco and put it as a healing plant. There‘s other things
to be considered I think
Vera: Well it‘s the pollen and the seeds isn‘t it
Sofia: It is
RM: Are you worried about it spreading?
Vera: Yeah
Molly: And animals eating it, if the grass is affected and then there‘s the sheep or the
cows or whatever
Sofia: Yes, there‘s a lot of things to think on
Elsie: They‘d have to be grown under cover wouldn‘t they?
Sofia: In a greenhouse, in a greenhouse, they will, they‘ll have to be grown in
greenhouses. If it was grown in a field, I, I don‘t think that‘d be safe
(Group D, meeting 2)

Group D here considers the role of place in the promise of biopharming. Their main
concern is that biopharmed crops will spread through pollen and seeds, and may
contaminate the environment, presenting a risk to animals and by extension to food. This
highlights how agricultural production is embedded in its surroundings and entwined in
networks of production. The dangers of relations with food production are elaborated as
Group E continues their discussion of biopharming:

Paul: If you‘re trying to grow these in food crops, or anything like food crops
because you don‘t know what folk will eat these days, I think that is a concern
Elizabeth: And I think you should pick your host so there‘s, if there is cross-
pollination, it‘s less likely to be with a food crop.
Paul: Yeah
Jane: And I would have thought the road to go down would‘ve been to grow these
things in a very controlled situation… And then you‘re looking at scale aren‘t you,
whether that‘s feasible, if you‘re going to have to grow large amounts

258
Helen: You don‘t want polytunnels everywhere
(Group E, meeting 2)

The choice of food crops, notably maize, is central to elaborating and contesting the
promise of biopharming. While for experts, using maize allows them to draw on existing
expertise and guarantees the safety of the plant vector, for Group E food crops are
problematic. Consequently, the definition of the places of biopharming is driven by the
risks of pharmaceutical crops to food and the environment.

However, while ‗placing‘ biopharming separates food from pharmaceuticals, the


polytunnels described by Jane and Helen, are still places of food production. In performing
containment, biopharming becomes linked to particular locations of production and
consequently to particular types of agriculture, to greenhouses and polytunnels. However,
for Group E, polytunnels are not simply a means of containment, but a structure that is
unwanted. As this example highlights, these places carry meaning, values and identity for
the practices that occur within them. Similarly, in the discussion from Group F below,
production inside represents a particular type of farming:

Josh: What‘s stopping them though from keeping it inside, if they're doing it inside,
what‘s stopping them from doing that?
Max: It‘s cost isn't it?
Sam: Doing what?
Josh: Keeping, growing these things inside rather than getting...
Sam: Because you could only grow so much
Rebecca: But could you then grow it more times, like twice a year
Sam: It‘s obviously lots cheaper to have a field than it is to have a factory isn't it?
Especially the size of it
(Group F, meeting 2)

For Josh, growing pharmaceutical crops inside is the obvious and preferred option.
However, Sam and Max introduce the challenges to farming and biopharming presented by
changing the location of production. They draw on the characteristics of the type of
agriculture suggested by greenhouse or polytunnel production to argue that the financial
and spatial restrictions of greenhouse cultivation favour open field production. However, as
Rebecca introduces, and as the following extract from Group A expands, these discussions
259
contribute to the fragmentation of the agricultural geographies underpinning
pharmaceutical production:

Rob: Could you imagine, if you had to, if you wanted to mass produce lipase, this
plant that produces lipase, you'd have to have huge fields, and massive greenhouses,
and that would mean you'd have to have all the like complex watering systems,
something to reduce the heat because greenhouses are going to get really hot in the
summer, there'd be like, you could imagine there'd be huge costs involved which
would impact on the environment, perhaps worse than having it as an open field
Will: But that already exists though, doesn't it, in terms of sort of daffodil production,
in Holland for instance
(Group A, meeting 2)

This extract links the imagined geographies of biopharming to the practical realities of
farming PMPs. The ‗hype‘ of the earlier discussion turns to pragmatic consideration of the
minutiae of production. Rob describes the challenge presented by the constraints of
greenhouse production and whether such production is desirable. He draws on an
understanding of existing greenhouse production, as does Will in contesting his
problematisation of biopharming.

Existing uses of greenhouses such as those described by Group A are also described in
other groups. For example Group F, continuing their discussion of the feasibility of
containment, discuss strawberry and tomato production in support of greenhouse
production:

Rebecca: It would be weird to have them all growing inside; it would have to be a big
greenhouse
Katie: Don't they do that in Spain with tomatoes
Sam: I think all tomatoes, or not all, I think a lot of tomatoes are grown inside
Katie: So you get them all year round
Sam: And strawberries now I think, or am I making that up?
Max: No you're right
Katie: Really like giant greenhouses
(Group F, meeting 2)

Greenhouses are normalised by association with existing spaces of food production. In both
discussions, biopharming is transformed from an abstract technology growing in
260
unspecified fields for large scale imagined applications to a real technology situated in
specific and meaningful places, the greenhouses of Holland and Spain. The restrictions that
greenhouses place on agricultural space are surmounted by evoking a different type of
agriculture. However, in the process the imaginary which supports the development of
biopharming changes. The technology itself is transformed from global and abstract, to
local and situated in the greenhouses of Spain or Holland.

For focus group participants, the purpose of greenhouse containment would be to separate
the spaces of pharmaceuticals and foods. However, in becoming concrete (or glass) in this
way, discursive relations with the places of food production are re-established, and
participants are able to draw on their extensive knowledge of these places. Greenhouse
production is related to other closed agricultural production facilities. Despite the arable
nature of biopharming, the most prominent sites of comparison are battery farms, discussed
in five groups: A, B, C, E and F. However, discussion of battery farming and factory based
food production emphasises the closed and invisible nature of production which prevents
groups effectively assessing the quality of food production.

A number of groups develop discussion of biopharming by drawing on the example of an


outbreak of avian influenza on a UK turkey farm. In early 2007, when the focus groups
met, the British news was dominated by an outbreak of H5N1 avian influenza at an
ostensibly sealed turkey farm run by Bernard Matthews in Suffolk. The question of
containment, or its failure, was central to the question of how the Suffolk birds had caught
the disease in a supposedly sealed food production facility.

While the focus group conversations in the first meeting emphasised how factory farming
and food production prevented the assurance of food quality, in the context of biopharming
it is on the difficulties of ensuring such places are effectively enclosed. As with the
example of ‗Terminator Technologies‘, biopharming leads to a public re-framing of closed
and restricted spaces and places of food. The role of avian influenza in discussion is
illustrated in the following extract from Group E:

Paul: This seems to be greenhouse grown doesn‘t it?


Jane: Yes, I suppose so, it‘s all under cover and controlled,
261
Paul: You could theoretically seal it, I mean even if your whitefly or your greenfly got
in, you could stop their predators from taking it further
Douglas: You could keep it at negative pressure couldn‘t you, and then filter the air
coming out
Alan: But that was supposed to be done though with the turkey farm wasn‘t it?
Helen: It didn‘t work then
Alan: And it didn‘t work there, you know, with this bird flu so…
(Group E, meeting 2)

In this discussion the ‗turkey farm‘ is combined with participants‘ own knowledges of
gardening and greenhouses to provide a repertoire through which groups consider
containment facilities. This repertoire functions not only to place biopharming, but as a
cautionary tale, even for greenhouse security above and beyond that of the everyday
glasshouse. Uncertainty is introduced into the places of biopharming as the H5N1 episode
questions the feasibility of physical containment. For example, in the quotation below,
Group B work through proposed containment options, but unlike other groups, have up to
that point ignored greenhouses. When prompted, they place greenhouses equally
questionable to other rejected methods:

Rita: It‘s not foolproof, look at the turkeys


Jo: Someone could walk out with something on their…
Rita: Human error, carelessness, you know
Sarah: And the thing about greenhouses, I mean similar to the turkey thing as well, is
that if for some reason something does, you know, get you know, it does get cross-
pollinated or whatever, then that whole batch is out as a result, and that‘s your
disaster isn‘t it, your whole crop is gone because of one error
(Group B, meeting 2)

The H5N1 discussion highlights that physical containment is not about barriers alone, but
in order to succeed involves disciplining fallible human actors. The challenge to physical
containment comes not only from the failure of the buildings themselves, but from the
humans within them. While the emphasis throughout these discussions is on the role of
greenhouses in protecting the food supply and the environment, this quotation introduces a
threat to the crops within them. As at the turkey farm, where 159,000 birds had to be

262
slaughtered after becoming infected24, it is pharmaceutical plants that are at risk from
contamination.

Publics consider contained production of biopharming as inevitably involving restrictions


on the production scale of biopharming. However, reference to alternative agricultural
systems re-establishes the feasibility of biopharming in general, placing it as equivalent to
European glasshouse farming. The link with existing places of farming allows groups to
develop an assessment of the characteristics of biopharmaceutical place that is not seen in
earlier group discussions, and which draws on widely shared mediated images of
biosecurity and containment practices. In contrast to the discussion of the same places in
the context of food production, closed production is considered in purely practical terms,
with less emphasis on the concealed nature of production.

6.3.2 Experts: Greenhouses and GMP

While publics emphasise relations with food production, expert discussion of the places of
biopharming is shaped by a two-way relationship between agricultural and biomedical
space. The need to protect biopharmaceutical agriculture from contamination is combined
with that of equating GMP quality guidelines. For experts, enclosed production excludes
biopharming from agricultural space both physically and symbolically. Contained
production separates pharmaceutical crops from fields and from food agriculture, but also
from food and agricultural regulation. As Commandeur and Twyman (2004) describe,
separating biopharming from food agriculture is a question of separating the ‗facilities‘ in
which each occurs:

―Ideally, there should be a clear distinction between transgenic plant material used
for molecular farming and any normal plant material being processed in the same
facility, which is intended for human or domestic animal consumption. A rigorous
series of regulatory practices should be in place from the farm to the factory,
ensuring complete isolation of transgenic material during growth, harvesting,
transport, storage, processing, extraction and waste disposal‖
(Commandeur and Twyman 2004:264)

24
http://news.bbc.co.uk/1/hi/uk/6356007.stm
263
Combating the problem of contamination requires the separation of the sites, facilities and
flows of food and pharmaceuticals. Successful containment of plant made pharmaceuticals
thus involves the development of a parallel farming system, growing food crops such as
maize yet separated at every stage from food production. Such a system is evoked in
researcher descriptions of the future facilities of biopharming:

I could very much imagine a big farm setting containing transgenic plants,
pharmaceutical production plants and then maybe in the midst of all those fields
having a processing facility where the whole harvest comes together, and then
around it a reasonable zone where no maize is grown for example.
(Dr H.)

This extract introduces how a separation between food and non-food agriculture can be
established. Despite the terminology of farms, plants and harvests, this system is isolated
and separated from food production by a maize-free zone. As explored in another
interview, this control of the sites at which biopharming occurs separates it from
conventional agriculture:

Professor C.: These crops are not really for regular farmers, they're for dedicated
facilities, and rather than seeing these things produced on a normal kind of farm, the
way it‘s going now is the company producing, wanting to produce them, will buy an
area of land, they will kind of fence it off, and they will control everything, and, so,
this would all, it‘s not really, as I say is not really something that would change the
profitability of farming across the board
RM: So in many ways, these crops are almost no longer agricultural are they?
Professor C.: I think so, that's how I envisage it now, that they, it will be a kind of
production unit rather than another crop to put into a regular agricultural rotation.
(Professor C.)

The scale and containment of production separate it from conventional agriculture, not only
spatially but in terms of the profitability of production to farmers. Rather than being
‗another crop‘, biopharmed crops are ‗production units‘. Separation dominates expert
discussion of the link with agricultural space and protecting the integrity of food
production.

264
The approaches described by researchers C. and H. target the separation of biopharming
from agriculture by distinguishing agricultural from pharmaceutical space. Greenhouses are
the most prominent of the diverse ways of establishing this separation. Greenhouses allow
experts to exclude biopharming from the physical spaces of agriculture. At the same time,
they remove it from the European regulatory space of food and agricultural biotechnology.
As the 2008 EFSA guidance document on biopharming states, ―the use of GM plants under
contained conditions (for example in greenhouses) does not fall within the remit of EFSA‖
(EFSA, 2008: 6). Instead, it falls under the ‗contained use‘ regulations laid out in
Regulation 90/220/EC and thus individual competent authorities. As one researcher
describes, this forms part of the calculations involved in the PPC‘s decision to grow its
crops in greenhouses, as:

―It‘s much easier to do it in greenhouses, because it just cuts out all the hassle of
getting all the permissions to do field trials‖
(Professor B.)

In both material and regulatory terms, contained production represents an effective break
between biopharming and agriculture. In addition, they allow PMP production to equate the
regulatory and material spaces of medicine. Expert concern about the contamination of the
environment and the food supply is accompanied by concern about achieving regulatory
standards of pharmaceutical production, particularly GMP. As Professor C. describes in the
extract below, a key advantage of greenhouses is that they enable the application of
pharmaceutical GMP to agricultural processes:

The bit in the middle, the agricultural bit, I think we need to define GMP more from
first principles there, because if you've got things in a fermenter you can control
everything that goes in, you can control the environment with some degree of
accuracy and so you can develop standard operating practices rather more easily. So
I think with the cultivation bit, that's the bit we need to.., that's where the real
challenges are. Now we've decided in PPC, for the life of PPC to work in
containment, so we'll be in a contained glasshouse, which is rather easier than
putting them outside
(Professor C.)

265
For Professor C. the glasshouse provides at least a short term solution to the challenge of
cGMP. Contained production allows some of the characteristics of the fermenter to be
brought to PMP farming. However, as described by publics, the containment of
biopharming within greenhouses has significant consequences for the development of the
technology and for understandings of its promise.

Contained greenhouses are co-produced with biopharming and understandings of its


potential, particularly the limitation of this potential. One review of molecular farming
describes how greenhouse containment ―negates the advantages of the technology in terms
of low cost and large-scale production‖ (Ma et al., 2003: 803), as confirmed in an interview
with Dr J.:

It‘s always said what you need is sun, water and a bit of soil, that‘s it. But this is not
true of course, and if for reasons of regulatory concerns for instance, you are forced
to go into the greenhouse, then of course you have quite different costs, electricity,
the building itself, and so it‘s hard to really find a system currently which is
competitive
(Dr J.)

While greenhouse production has the advantage of meeting pharmaceutical regulatory


concerns, it conflicts with the long term goals and spatial imaginaries of biopharming. One
of the key benefits of a plant based production system is its use of agricultural scale and the
production of pharmaceuticals in quantities impossible in the confined bioreactors of
conventional biopharmaceutical manufacture. In greenhouses, the simplicity of agricultural
space, and the low-tech nature of production are spatially restricted. By equating bioreactor
conditions and excluding agriculture, the agricultural advantages of the technology are lost.
As Professor C. continues:

It‘s always argued that the advantage of plant produced pharmaceuticals is that you
can scale up relatively easily, all you need is a bit more land, you've got the machine,
you've got all the tackle, and whether you grow 10 acres or 50 acres, it‘s just a bit
more labour, you need more land, but it‘s not double the cost or five times the cost,
it‘s probably only half as much again in terms of production. So, that is a principle
advantage of growing stuff outside. If you do it in a glasshouse then, you know,
glasshouses can be quite expensive

266
(Professor C.)

The promise of biopharming is located in its specific places and scales of production.
Moving from fields to indoor production compromises the very advantages that distinguish
biopharming from existing pharmaceutical manufacture. Greenhouse production removes
the easy scalability of production which separates fields of pharmaceutical crops from large
steel bioreactors. However, in further interviews and in research publications, promise is
renegotiated through reference to a new agricultural space:

Even under containment, it would be possible to grow a large number of


pharmaceutical plants; immense greenhouse facilities are already used routinely by
the horticultural and food industries.
(Ma, Barros et al., 2005: 594)

While the purpose of physical containment in the extracts above relates to achieving
pharmaceutical containment, the establishment of GMP is here seen as a step on the road to
field production. In thinking about the longer term production of pharmaceuticals,
containment again becomes a question of environmental separation. The feasibility of long
term containment is affirmed through its similarity to other sites of agriculture, different
from those which problematise pharmaceutical spaces.

Agricultural space as messy and unruly is nuanced into two different types of sites: those of
open agricultural production and those of ‗immense greenhouse facilities‘. This represents a
redefinition of the initial association of molecular farming with agricultural production,
instead associating it with particular types and places of agriculture. From representing
potential problems for molecular farming requiring the use of enclosed production space,
existing agriculture now underpins the potential competitiveness of molecular farming. In
particular, greenhouses are still competitive with existing technology.

What you say is you're trading fermenter capacity for greenhouse space, that‘s the
point really, and so where you're saving is on growing up the biomass. You're not
saving on GMP processing because the processing is tricky and expensive anyway, it
is expensive in fermenting, it‘s expensive with plant material, but it‘s the fact that a
greenhouse is easier to build and easier to maintain and it‘s cheaper to build than a
fermenter

267
(Dr G.)

Greenhouses and biofermenters are here found to be comparable approaches to the part of
pharmaceutical production which is flexible, or ‗upstream‘ processes. Rather than being a
potential hindrance, greenhouses as a contained, safe space now become part of molecular
farming‘s promise. The possibility and attractiveness of greenhouse production is boosted
by the reintroduction and elaboration of a relationship with agricultural spaces, as
researchers draw on existing large scale production in greenhouses.

In the discussions above, greenhouses become both pharmaceutical and agricultural


production spaces. However, the agriculture discussed is no longer that of conventional
farming or of first generation agricultural biotechnology. Nor is it any longer abstract,
placeless agricultural space, but becomes concrete and located in existing production, as
described in an interview with Professor D.:

So I mean, the two plants that are talked of [for edible vaccines], I mean talked of
seriously, are lettuce and tomato, and both of those at least in Europe tend to be
grown under cover anyway. If you look at where the lettuce comes from, it comes
from Holland and it‘s all grown in greenhouses, so then you haven't got the problem.
I mean you might have the problem of mixing things up, but obviously you can do
that. The question is then, most of these things part of the reason for doing it is the
Third World, so the question is then how do you do it, do you then have to grow them
in the third world, and my view is probably not.
(Professor D.)

Dutch greenhouse production of lettuces reinforces the potential of biopharming. Similarly,


in a quote introduced to focus groups, Ma, Barros et al. suggest that ―250 acres of
greenhouse space would be sufficient to grow enough transgenic potato plants to meet
South East Asia‘s annual demand for the hepatitis B virus vaccine‖ (Ma, Barros et al. 2005:
594). However, the use of greenhouses opens up a new rift with the aims of molecular
farming. While the use of greenhouses is justified through reference to existing agricultural
production, and the scale of production is re-imagined, greenhouse production is itself
located. The concentration of greenhouse production in Europe and the United States
means that molecular farming facilities becomes linked to these places and separated from
the original goals of research. Rather than the imagined open, global spaces of agricultural
268
production which support the development of the technology, the places of molecular
farming become finite and physically immobile within national and international contexts
of agricultural production and circulation. Daffodil and lettuce production in these two
discussions are not 'placeless' parts of globalised agri-industrial production, but are sited,
grounded in Holland.

The move to greenhouses achieves more than protect the integrity of the food supply. It
allows the combination of both agricultural and pharmaceutical concerns. To become
pharmaceutical, molecular farming must achieve equivalence to the quality regimes of
existing pharmaceuticals. As for the stem cells described by Franklin (2006) and Stephens
et al. (2008), quality assurance is located in biomedical production sites. The large steel
bioreactors containing bacteria or Chinese Hamster Ovary (CHO) cells which produce
existing biopharmaceuticals provide a highly regulated and controlled environment. The
use of a greenhouse reduces the complexity of agricultural production and allows the
establishment of this control. It enables a micro-control over inputs and outputs that is
impossible in conventional farming. However, the combination of these two drives to
contain biopharming entails a reconsideration of the places and promise of the technology.

6.4 Resolving the Places of Biopharming,

I have considered how the placing of biopharming through physical containment is shaped
by the need to assure the quality and safety of both agricultural and pharmaceutical
production. In bringing this chapter and the thesis towards a close, I examine how this
results in the emerging ‗places‘ of PPC biopharming.

Physically enclosed production excludes biopharming from agricultural space both


physically and symbolically. In addition, it allows the control required for pharmaceutical
production to be added to agricultural production. For experts and publics the potential of
greenhouse production of biopharming is supported by examples of existing production.
However, the spaces of agricultural and pharmaceutical greenhouse production are not
currently the same.

269
Figure 6.2 Greenhouses at Fraunhofer IME, Aachen

The greenhouses shown in Figure 6.2, located next to the Fraunhofer Institute for
Molecular Biology and Applied Ecology building on the academic campus of RWTH
Aachen, Germany, are one of the key research sites for the development of maize based
production. The IME is one of 57 Fraunhofer Institutes across Germany, intended to
produce ―applied research … of direct utility to private and public enterprise‖25, and is one
of four in Aachen, at a university founded to educate engineers for the Ruhr valley mining
industry. The Fraunhofer Institutes are boundary organisations between public and private
sector research, and the location and buildings of Fraunhofer IME reflect its close links
with both the German biotechnology industry and academic biotechnology research in
other parts of the campus. The landscaped gardens and architecture highlight that this is a
modern biotechnology facility.

These greenhouses are closed and contained. Filters on the top of the buildings control
movement in and out, as for the GMP building. Inside, the maize plants are grown on
benches, preventing run-off from the plants and allowing control of watering. The

25
http://www.fraunhofer.de/EN/company/mission/index.jsp
270
restrictions of greenhouse production described above are evident, as the height of the
maize plants is limited by the greenhouse ceilings. Nevertheless, greenhouse spaces
depicted in this picture still lack many of the tightly controlled characteristics described in
discussion of GMP facilities. As one interviewee introduces, this demand erodes the
potential of biopharming:

RM: So you think in order to meet EMEA requirements, molecular farming is going
to have to take place in greenhouses?
Dr J.: Yes, this is the current state of affairs. I think that was my impression. Which
cuts your potential savings in half. In the EMEA draft plans they even said regular
greenhouse design might not be adequate, they wanted advanced greenhouse design,
I don't know a high security area or something, which is comparable to the costs of a
fermenter
(Dr J.)

The importance of achieving acceptable pharmaceutical space is potentially crippling for


biopharming, and the agricultural imaginary of researchers, which has already moved from
open fields to agricultural greenhouses, is further challenged. Dr J.‘s description of the
EMEA guidance refers to their 2002 ‗points to consider‘, which stated that:

"ordinary greenhouse conditions do not provide a high level of protection...advanced


greenhouse designs may offer more protection"
(EMEA 2002:6)

The EU regulatory framework for biopharming has evolved since then, up to the
publication of the EFSA draft opinion and EMEA‘s final guidelines in the summer of 2008.
The requirement for advanced greenhouse design was dropped from subsequent EMEA
guidelines. However, as Dr J. suggests, this requirement continues to play a role in
researcher discussion of production, and is reflected in the form taken by actual greenhouse
production of PMPs by PPC researchers in Kent, as described in a 2006 newspaper report:

―The plants are being grown in £35,000 high-security Unigro greenhouses which
normally house experiments on plant viruses. Designed to withstand a lobbed brick,
the greenhouses are twin-skinned plastic. Rupture either skin and the entire
greenhouse is immediately flooded with formaldehyde, keeping everything inside‖
(Sample 2006)

271
This last movement to a Unigro Grodome in Kent represents the final step in the
containment driven move from open agricultural spaces filled with excreta and parts of
animal carcasses, through the lettuce greenhouses of Holland to a facility featuring:

―a microbial deactivation waste system...an air pressure control system; filtered


venting; pressurised sealed doors; personnel activated exit and entry systems and 36
hour uninterrupted environmental conditions in the event of mechanical failure‖
(www.unigro.com)

The Unigro facilities provide high levels of containment in agricultural settings. Figure 6.3
is illustrative of how this containment is ensured. As are GMP facilities, it is a standardised
facility which can be effectively replicated.

Figure 6.3 The pressurised corridor leading to research chambers in the Unigro
Grodome (from www.unigro.com)

The use of the Unigro system to equate pharmaceutical biofermenters has been funded by
the UK Department of Trade and Industry26, as the promise of biopharming shifts back into
the conventional economic as well as physical spaces of pharmaceutical manufacture.

26
http://www.unigrow.co.uk/pages/news.html
272
Equating the quality demands of pharmaceutical regulation has driven expert
understandings and enactments of the places of biopharming.

However, given that the requirement for ‗advanced greenhouse design‘ has been dropped
from EMEA guidelines, equating GMP alone may not be enough to explain the
characteristics of these spaces. Instead, the protection of the environment and food supply
re-emerges in the GroDome. From the flooding of the facility with formaldehyde in the
event of a breach of containment, to Sample‘s description of it as resistant to a ‗lobbed
brick‘, the GroDome re-establishes a link between the places of biopharming, the
characteristics of its products and its imaginaries. Despite the pharmaceutical
characteristics of the environment, its form is in part dictated by its agricultural
biotechnology heritage and the openness of fields and glass buildings to the protest and
‗ecotage‘ that characterised the places of first generation GM. Together, the agricultural
and pharmaceutical ‗placings‘ of biopharming result in the emergence of ‗hybrid places‘
which combine characteristics of agricultural and pharmaceutical space.

6.5 Summary and Conclusions

Exploring the process of ‗placing‘ offers a productive approach to considering the diverse
ways in which new technologies enter into relations with existing ones. I have described
how this occurs through the association of biopharming with the narratives and materials of
food and medicine. I have now considered this process is completed through the association
of biopharming with the material places of pharmaceuticals and foods. The discussion is
summarised in Figure 6.4.

I describe how the promise of biopharming moves from the imagined geographies of the
developing world and large-scale agriculture to be located among the geographies of
Western glasshouse agriculture and contained pharmaceutical production. This movement
is driven by the characteristics of both agricultural and pharmaceutical production. In
section 6.1 I described how the places of food play an essential role in the establishment of
food quality in what Murdoch (2005) describes as ‗topological‘ networks. The spatial
origins of food production are linked to judgements of the production process. For experts,
273
it is pharmaceuticals whose quality is guaranteed by their places of production. Achieving
pharmaceutical Good Manufacturing Practice (GMP) is bound up with particular facilities
and ‗performative architectures‘ (Stephens et al. 2008).

Figure 6.4 Summary of Chapter Six

Agriculture Pharmaceuticals

The places of food contribute to assuring food Pharmaceuticals are placeless – the
quality and safety, particularly when they geographies of their manufacture and
allow production to be witnessed consumption are obscured by practices of
testing and the authority of expertise
The open spaces of agriculture are threatened
Publics

by the pharmaceutical products of biopharming The topologies of pharmaceutical


manufacture are made visible by
Containment and the adoption of alternative controversies
agricultural space is required
However, biopharming is different to
existing pharmaceutical space due to its
relations with agriculture

Biopharming threatens agricultural space, but Pharmaceutical places, and the control and
this threat is perceived as much as real – not all visibility of production within them, assure
biopharming presents the same threat pharmaceutical qualities
Experts

Existing greenhouse agriculture is used to Equating the characteristics of regulation-


reassert the promise of biopharming in the face approved, GMP controlled pharmaceutical
of requirements for containment space becomes the key driver in the
development of contained places for
biopharming.

Biopharming represents a challenge to both of these located quality regimes. For publics,
enthusiasm for the promise of biopharming is predicated on the withdrawal of parts of
agricultural space from food production and agriculture in general. Biopharmaceutical
production represents a threat to the integrity of food agriculture and to the environment.
For experts, particular PMPs, those which are ‗pharmaceutically active‘, are also
threatening to food production.

274
However, for PPC experts, agricultural production space is in itself threatening to the
quality of pharmaceutical products and the ability to equate GMP production. Agriculture is
unruly and messy – everything that pharmaceutical production facilities are not.

The need to separate pharmaceutical and food production, as well as products, leads
researchers and publics to focus on containment or confinement as necessary requirements
for biopharming. Greenhouses are foremost among the means to do this. However, to
become feasible production environments they require a discursive realignment of
biopharming with European glasshouse farming of tomatoes, daffodils and lettuces, rather
than open agricultural spaces. Yet these spaces themselves are still not ‗pharmaceutical‘.
Instead, a final movement into hybrid places that combine agriculture with the control of
pharmaceutical space is required.

In the final chapter, I bring this discussion together with that of Chapters Four and Five to
consider how the temporal, ontological and spatial placings of biopharming combine in
understanding the formation and evolution of technological promise.

275
CHAPTER 7: NO PLACE LIKE HOME

Work in science and technology studies emphasises that new science and technology is a
'heterogeneous assemblage' of people and things (Latour 2005; Bijker 1995).
Technoscientific change and the successful introduction of a new technology involves the
formation and stabilisation of new socio-technical associations. One means through which
this is achieved is by the production and circulation of promises or visions of technological
futures. These visions enrol publics in 'communities of promise' and shape the technical
choices made by researchers. This is particularly evident in the case of biotechnology. Both
agricultural and pharmaceutical applications of biotechnology have been characterised by
their attempts to colonise the future and imagine worlds transformed by their introduction.
Yet the early promises of red and green biotechnology were challenged by a range of
social, technical and geographical factors, giving rise to new visions, new futures and new
technologies. Among these new technologies is the production of pharmaceuticals in
genetically modified crops, or biopharming.

In this thesis I set out to examine expert and public perspectives on the development of
biopharming. I have described how experts attempt to stabilise and materialise the products
and places of biopharming, establishing them as pharmaceutical while retaining the
agricultural features that make the technology so promising. I have also discussed how
publics, while enrolled in the promise of biopharming, focus on its relations with food and
agriculture and the threat of contamination of food supplies. Through both sites, I have
explored how the promise of biopharming is performed through its relations with existing
green and red technologies. Bringing the geographical metaphors of STS to the fore, and
combining them with work that explores the geographies of science, I have sought to
explore the heterogeneous ways in which these relations are established through the
heuristic of ‗placing‘.

276
In this chapter I first summarise the conclusions of the preceding chapters, bringing them
together with these perspectives to suggest that together they effectively depict the
heterogeneous ways through which biopharming is co-produced with new social, technical
and geographical orderings. In section 7.2 I consider the contributions of my research to
these broader discussions within STS. In particular, I consider the ways in which the thesis
contributes to understanding the geographies of technological expectations. I then discuss
potential limitations and concerns about the research strategy adopted here and the
depiction of biopharming it presents. Finally, I suggest that the example of biopharming
opens up new directions for research, in considering the geographies of both technological
expectations and of pharmaceutical production more generally.

7.1 Summary and Synthesis

Throughout this thesis, I have described how biopharming is produced through its
associations with (and dissociations from) the characteristics of agriculture and
pharmaceutical production. I introduced this in terms of the 'placing' of biopharming - a
process through which its locations, contexts and meanings are established. The heuristic of
placing brings together three interlinked notions of place to explore the relations and
tensions between them. My discussion breaks these down into public and export efforts to
‗date‘, ‗classify‘ and ‗locate‘ biopharming in the context of existing technologies. It
contributes to understanding how ‗placing‘ technological promise is a process of narration,
but also of the establishment of material and spatial continuities and discontinuities.

The circulation of expectations constitutes a ‗place‘ in time for new technologies,


positioning them in relation to their future, but also their past. In his analysis of beginnings
in literature, Edward Said (1975) describes their importance in associating new endeavours
with the 'authority of a parallel venture'. In my empirical discussion in Chapter Four, I
examined how the narration of biopharming‘s pasts in terms of pharmaceutical beginnings
contributes to the formation of a ‗community of promise‘ around biopharming. For publics,
the pasts of penicillin and of vaccination demonstrate past success in biomedical

277
innovation. The unpredictable, serendipitous nature of these successes supports the
potential of biopharming and contributes to the forcefulness of biomedical futures.

Biopharming is also placed among the imagined spaces, materials and practices of
developing world agriculture. This contributes not only to establishing biopharming‘s
‗symbolic capital‘ (cf. Rajan 2006), but to shaping technological choices. In particular, the
selection of maize as the production platform for the PPC is based on its characteristics as a
low-tech stable platform, rather than as a foodstuff. Maize and the characteristics of food
are central to the discussion of Chapter Five, in which I examine how the materials of
biopharming are classified and ‗placed‘ among those of food and medicine. The products of
technoscience come to embody particular visions and expectations of their future.
Expectations of biopharming are supported by the association of its novel materialities with
existing ones. However, these materials are also the means through which biopharming is
encountered and its meanings challenged.

The discussion of maize and glycosylation highlights how the materials of foods and
pharmaceuticals act to influence the ability of biopharming to effectively colonise the
futures mapped out in the narrative accounts. However, it also demonstrates that the
meanings attributed to these materialities are contingent, and the relevance of their
obduracy is constructed . For example, in the case of maize, it highlights that the same
materiality can be used to enable and challenge biopharming‘s promise. While the food
characteristics of maize allow biopharming to draw on a ‗history of safe consumption‘,
other plant characteristics such as glycosylation challenge its pharmaceutical safety. In the
case of edible vaccines, the same qualities that convince some experts and publics of the
ease of delivery are challenged by the too-close biological and social relations between
vaccines and food.

A central way through which encounters with the materials of biopharming are staged is
through their relationship to a variety of ‗natures‘. Throughout the development of
biotechnologies, their relation to the 'natural' and the 'unnatural' has been a central problem,
but there is no simple relation between ideas of nature and the practices of and attitudes to

278
biopharming. For concerned consumers, nature is used to establish the authenticity of
foods, and to ensure a minimum of processing. However, it also contributes to the
classification of food described above. Similarly, for experts, nature is the standard against
which plant made pharmaceuticals are judged – both in terms of the safety of existing food
products, and in the form of ‗natural‘ patterns of protein glycosylation.

I closed Chapter Five considering attempts to reinstate material distinctions between food
and pharmaceuticals through ‗identity preservation‘. In Chapter Six, I expanded this
discussion to examine how concerns about encounters between the materials and places of
pharmaceutical and food production prompt their separation. I described the importance of
the places of food production in consumer discussion of food quality. Closed and concealed
places are associated with poor quality food, while the places that are valued are those in
which production and preparation can be observed. This close link between quality and
place emphasises that in describing focus group approaches to food, the separation of food
product and production process is difficult.

Focus group discussion of food provides an interesting parallel to expert discussion of


biopharmaceuticals. For publics, pharmaceutical production is black-boxed, and is instead
validated and verified by trusted intermediaries. As in the case of food scares, this box is
opened by controversies, such as the failure of the TGN1412 clinical trials. In contrast, for
experts the quality and safety of biological medicines is inextricable from their production
– ‗the process is the product‘ – and consequently from the places in which this happens.

However, biopharming – and in particular maize biopharming - represents challenges to


agricultural and pharmaceutical geographies. The danger of contamination of foods, the
environment and the pharmaceutical products drive the framing of the technology in terms
of containment. The separation of biopharming from agriculture by containment, and the
construction of new places of biopharming, involves revisiting its imagined geographies
and replacing open, developing world field production with developed world greenhouses.
As one set of imagined geographies is replaced by another, so the promise of the
technology is recreated.

279
The discussion of Chapter Six represents a move away from the discussions of narrative
and materiality in existing work on technological promise and expectations. The description
of place as ‗social location‘ in much existing work on expectations, although valuable,
neglects the role of the physical locations described in recent work on the geographies of
science. Having considered the multiple means through which a 'natural home' for
biopharming is constructed and its promise established and challenged, it is possible to
offer some conclusions on the relationship of biopharming to red and green
biotechnologies.

The first way to conceptualise this placing might be to describe biopharming as 'between'
red and green biotechnologies. This would suggest that these represent two existing poles
between which new biotechnologies are distributed. However, as discussion of the
pharmaceutical storage qualities of maize, of relations between developing world medicine
and farming, or of greenhouses and GMP suggests, the relevant qualities of agricultural and
pharmaceutical production are co-produced with the potential futures of biopharming.

Focussing on expert perspectives, a second suggestion might be that biopharming


undergoes a process of coming to be pharmaceutical. Taking agricultural biotechnology as
a starting point, one might describe how the association of biopharming with hopeful
pharmaceutical visions, with human protein glycosylation and with GMP facilities
represents a linear movement from green to red biotechnology. This narrative is particularly
plausible when expert discussions of biopharming are considered. For the PPC, achieving a
pharmaceutical home for biopharming involves establishing the quality, safety and efficacy
of the pharmaceutical product.

Approaching biopharming from the perspective of focus groups, a parallel narrative runs
through the thesis of a process of ceasing to be food. Again, biopharming starts in
association with the hopeful pharmaceutical futures of penicillin and the geographies of
developing world healthcare. However, from a similar starting point, public discussion of
biopharming as we move through Chapters Five and Six emphasises associations not with
pharmaceuticals, but rather dissociations with the products and places of food and

280
agriculture. Focus group participants concentrate on establishing and maintaining material
and symbolic boundaries between pharmaceutical and food agricultures.

These three approaches suggest that 'placing' biopharming is a situated act - that any
placing is partial and located. Consequently, the promise of biopharming is enacted through
multiple geographies. The elaboration of these geographies, which I discuss in the next
section, forms the thesis's central contribution to work in science and technology studies
and to the geographies of science.

7.2 Exploring the Geographies of Biopharming

In this thesis, I have described a process of ‗placing‘ biopharming, in which it emerges


among ‗mundane and unremarkable networks‘ of established actors and materialities, yet
―de-stabilizes these networks in order to establish [its] own amenable and fertile
associations‖ (Brown and Michael 2003: 14).

The main contribution of this thesis is to expand discussion of the spaces of expectations.
My discussion of biopharming adds to this by describing the constitutive role of imagined
geographies in visions of the future. In addition, my thesis argues that these visions of the
future are challenged and shaped by the material and embodied geographies of foods and
pharmaceuticals.

Hopeful visions of the future have been a constant feature of new biotechnologies, from the
early development of biopharmaceuticals and agricultural applications described in Chapter
One, through to the case of biopharming. In Chapter Four, I suggested that expectations
incorporate not only imagined futures, but imagined geographies, and that these play an
important role in the formation of ‗communities of promise‘ around biopharming. I extend
this discussion below

281
7.2.1 Imagined Geographies of the Future

Within STS, network topologies have often dominated discussion of space. Geography has
too often been seen as context or scenery, rather than as playing an active role in the
development of novel technoscience. In recent years, there has been a turn towards
considering the geographies of technoscience in more detail. As I now describe, this can be
brought to bear to develop the discussion above.

The imagined geographies of developing world agriculture and medicine contribute to the
formation of ‗communities of promise‘ around biopharming. They provide a shared
representation around which public and expert visions for the future of the technology
come together. While existing work on ‗communities of promise‘ has tended to focus on
the enrolment of smaller groups of patients or of interested participants, to a large extent the
promise of biopharming is shared between experts and the wider, ‗disinterested‘ public
constituted by the focus group research.

The work of Benedict Anderson (1983) on ‗imagined communities‘ describes the


importance of vernacular language in the development of shared imaginations. In the case
of PPC biopharming, drawing on widely available discourses around HIV/AIDS brings
together public and expert understandings of the need for technological progress.
Moreover, the feasibility of biopharming‘s futures is established through an association
with past pharmaceutical innovation. Although this takes different forms, for example
whether vinblastine or penicillin is used as an example, the existence of authorising parallel
narratives makes the futures of biopharming credible.

However, public and expert visions of biopharming‘s future geographies do not entirely
converge. While expert discussion presents biopharming as a ‗technological fix‘ to diseases
such as HIV or rabies, public discussion emphasises its position among previous such
efforts. Here the wide availability of mediated images of HIV in the developing world
contributes to the emergence of sceptical perspectives and divergence between public and
expert expectations. In contrast, experts are unclear on how to resolve concerns about the

282
compatibility of salvational, placed visions of biopharming with the economic and
regulatory imperatives of European biopharmaceutical manufacture.

For Edward Said (1995) imagined geographies establish identity and authorise action in
relation to ‗others‘. In his discussion of Orientalism, he suggests that the colonial projects
of Europe were enabled by contrasting a civilised Europe with a view of the barbarian
Orient constructed through literary and artistic representations. While it would be
unreasonable to suggest that biopharming represents a spatial neo-colonialism, both public
and expert understandings of the technology are based on representations of the developing
world which enable or challenge the suitability of particular technological interventions. As
such, they enable the colonisation of socio-technical futures.

As are Said‘s imagined geographies of the Orient, those of biopharming are performative.
Visions of developing world agriculture and medicine shape the development of the
technology by the Pharma-Planta Consortium. Expert descriptions of the places, scales and
natures of agriculture are integral to the promise of biopharming and to supporting the
choice of maize as a pharmaceutical production platform. However, as a major food crop
which is wind-pollinated, maize constitutes particular symbolic and material associations
between biopharming, food and first generation agricultural biotechnologies.

Existing research in the sociology of expectations describes how visions such as those
described here, and the certainty with which they are expressed, are situated in both time
and space. Brown and Michael (2003) suggest that the stories that are told about
technoscientific futures are shaped by the times and places of the actors who formulate
them, through a situated process of ‗retrospecting prospects‘ and ‗prospecting retrospects‘.

Existing research has concentrated on the ‗social locations‘ of expectations and the socio-
spatial ‗distance‘ of actors from innovation. In Chapter Two, I suggested that these socio-
spatial distributions are those of ‗expecting‘ as much as expectations, and argued that
analysis can usefully be extended through an engagement with research in the geographies
of science. I discuss this work below, but first consider how my research on biopharming
contributes to understanding the social geographies of expecting. In particular, I am

283
interested in examining the relationship between publics and research in terms of the
distance from research, judgements of uncertainty and what Brown and Michael (2003)
describe as the future ‗naivety‘ that derives from distance from innovation.

7.2.2 Socio-Spatial Geographies of Expecting

In this section, I explore the socio-spatial distribution of expectations for biopharming. I


describe how the relational topologies of agriculture and medicine constitute different roles
for both experts and publics. I then suggest that the roles adopted by and attributed to
publics act to constitute ‗future naivety‘.

In Chapter One, I suggested that one product of my thesis would be a clearer understanding
of the relationship between the publics of agricultural and pharmaceutical biotechnology.
However, the research presented here suggests that direct comparison is problematic. By
putting the publics of agricultural biotechnology alongside those of pharmaceuticals in a
direct comparison of red and green, we posit an equivalence in their role and social
location. Moreover, the grounds for equivalence are shaped by the way it is approached.
Approaches such as that described by Marris et al. (2001) use public engagements with
agricultural biotechnology as a starting point, and discuss medical applications as a contrast
with genetically modified crops. Consequently, a list emerges that emphasises the points at
which pharmaceutical consumption differs from that of foods, based around questions of
choice, control and expertise.

In my research, encounters between publics and pharmaceuticals were again staged in the
same terms as for foods. Publics were asked to describe how and why they consumed
particular examples of over-the-counter medicines. However, my research suggests that in
the case of pharmaceuticals, the consumer role adopted by focus group participants in the
case of foods is, if not absent, significantly less pronounced. In the case of foods, reported
information and second-hand expertise are mistrusted in favour of direct experiential
assessment. In contrast, discussion of pharmaceuticals represents almost the inverse.

However, while direct comparison of the content of discussion is difficult, putting focus
group discussion as consumers of foods and medicines alongside each other suggests that
284
while practices differ, this is not necessarily because of a ‗red good/green bad‘ divide.
While some participants do emphasise the ideal characteristics described by Marris et al.,
they tend to be those whose ‗distance‘ from medicine is greatest. In contrast, participants
who are more familiar with medicines are more critical, both of the characteristics of
medicines themselves and of the experts responsible for their provision. Moreover, when
the characteristics of pharmaceuticals are brought into the realm of consumer knowledges
of food in the case of edible vaccines, the promise of biopharming is immediately
challenged and the complex discussion of the places of containment described in Chapter
Six emerges. Consequently, the value attributed to pharmaceutical information and
expertise may not necessarily be a question of choice, but rather an adaptation to the lack of
available avenues for the development and expression of consumer-citizen identities and
limitations on the acquisition of information and expertise about medicines.

The consequences of this for understanding ‗future naivety‘ and the formation of
‗communities of promise‘ are obvious. At the most basic level, they suggest that publics
occupy different roles in relation to agriculture and medicine, and consequently that
different technologies perform different publics. However, they also suggest that attempts
to involve wider publics as ‗active consumers‘ in biomedical innovation are problematic
given the undeveloped nature of this role.

While Brown (2007) suggests that the authorship of expectations is distributed among
'communities of promise', a contrast between agricultural and pharmaceutical
biotechnologies highlights that the constituency of authors is restricted by existing
relationships around food and medicines. This suggests a need to refine the 'socio-spatial'
geographies of expectations described above. In Chapter Two, I suggested that Benedict
Anderson's concept of 'imagined communities' can be criticised for the lack of emphasis on
whose imaginations are being circulated. Similarly in the case of expectations, over-
emphasising the role of lay actors in communities of promise conceals these restricted
constituencies and the asymmetries of power and knowledge that drive these
restrictions. Moreover, the possibility of effective resistance by ‗unimpressed publics and

285
professionals‘ (Brown and Michael 2003) is limited by the ability of, and opportunity for,
meaningful public engagement in pharmaceutical innovation.

In this thesis, I have described how the socio-spatial distributions of expectations described
above are complemented by imagined and material geographies. As I have described, the
distinction between these is not clear. Imagined geographies of maize agriculture and of the
developing world drive the development of particular types of biopharming, while new
physical locations, such as the Unigro greenhouse, involve the reworking of these imagined
geographies.

7.2.3 Obdurate Geographies

Finally, I focus on the role of the material geographies of technoscience in the performance
of futures. The case of biopharming demonstrates how the geographies of technoscience
shape both the content of technological promises and the constituency of those able to act
in regard to this promise.

Brown and Michael (2003) suggest that the success or failure of expectations is in part the
result of whether practical and material considerations ‗play along‘. As a ‗symmetrical
sociology of the future‘ the sociology of expectations asks ―how it is that language and
materiality are both, within given limitations, mutually constitutive of various futures‖
(Brown et al. 2000:15). In other words, narrative and material orderings are co-produced in
the emergence of technological futures. This symmetry or co-production can be observed as
imagined geographies of biopharming encounter and are shaped by the material
geographies of agriculture and medicine, and prompt the emergence of new materialities
and new places of novel technoscience.

Moreover, the case of biopharming highlights the interaction between these places and the
social topologies described above in shaping the authorship of technoscientific futures. The
work of Shapin (1988), Gieryn (2002) and Kohler (2003) shows how the places of science,
in particular laboratories, act to restrict those who are authorised to witness and attest the
reliability of knowledge. On the other hand, field sites involve ―a blurring of the boundary
between laboratory and world‖ (Szerszynski, 2005: 182) and the formation of new
286
relationships between the occupants of scientific and ‗social space‘ (Bonneuil et al. 2008).
The places of technoscience thus define the terms on which encounters between research
and publics occurs. As such they delineate possible relations between actors and
strengthening some associations at the expense of others. In the case of biopharming, it is
ultimately through these material geographies that ‗placing‘ is accomplished, and through
which relations between agriculture and medicine are stabilised.

The material placing of biopharming is dominated by discussion of containment. The


places and practices of containment provide a means of materialising relationships between
socio-technical assemblages of pharmaceuticals and agriculture. Placing biopharming in
contained greenhouses such as the Unigro ‗GroDome‘ restricts the movement of the
biologies of plant made pharmaceuticals and the dirt of agriculture. However, it also moves
biopharming from the open, visible sites of food production valued by focus group
participants as a proxy for quality, to the closed, controlled sites of pharmaceutical
production. The contained facilities of biopharming thus reverse the movement described
by Bonneuil et al. (2008) in the context of field trials, representing a withdrawal of
biotechnology from social space. Consequently containment involves the exclusion of
actors from that space and limitations on the extent of societal engagement.

As well as removing biopharming from ‗social space‘, containment also moves it into the
standardised, ‗topographical‘ (Murdoch 2005) spaces of pharmaceutical manufacture.
Stephens et al. (2008) and Franklin (2006) have described the assertion of biomedical
quality through particular places. The medical identity of the plants of biopharming is
established through regimes of good manufacturing practice. These control the material
associations of the new technology, while also establishing its position among
biopharmaceutical manufacturing practices. Together, these re-assert the pharmaceutical
promise of biopharming, but on significantly different terms to the ‗agricultural scales‘
described in Chapter Four.

287
7.3 Looking Beyond the Thesis

In the preceding sections I have described the contributions of this thesis to work in science
and technology studies, particularly focussing on its elaboration of a geography of
technological expectations. However, there are a number of obvious concerns to the
research, not least those that derive from the study of biopharming itself. In this section I
describe these concerns, and look beyond the narrow, contained world of the thesis to
consider its limitations.

The first set of concerns focus on the ability of the thesis to effectively represent
biopharming as a whole. As described in Chapter One and expanded in Chapter Three, the
discussion presented in this thesis concentrates on the work of the Pharma-Planta
Consortium and its vision for the development of biopharming. Yet as highlighted by
Brown, Rappert and Webster (2000) multiple competing visions of technologies are often
present during their early stages. Indeed, in my introduction to biopharming, I described
how US and Canadian companies such as Ventria Bioscience or Sembiosys are adopting
different approaches to the PPC, using different crops to target different molecules.
Moreover, the PPC is somewhat anomalous, not only in terms of biopharming, but in
pharmaceutical research more generally. As a publicly funded effort to enter a
pharmaceutical into clinical trials it is a rare sight in an industry dominated by large
companies or small biotechnology firms. The conclusions of this thesis are therefore
situated in the times and spaces in which research was conducted.

The decision to focus on the PPC was in part a pragmatic one in terms of the resources
available for a doctoral study. As an EU funded project, primarily based in academic
settings, the PPC provided an accessible focus for research. However, it also represented an
opportunity to focus on a large project which represents an attempt to ‗colonise‘ (Brown
and Michael 2003) the future of biopharming in a particular way, through establishing it as
a means of developing world pharmaceutical production. Furthermore, the PPC‘s remit
includes dealing with social and regulatory questions around biopharming as well as the

288
development of a new product. As such, it provided an excellent lens through which to
focus biopharming‘s relations with agriculture and medicine.

Nevertheless, given the divergence in national framings of agricultural and medical


biotechnologies described by Jasanoff (2005) a valuable comparison could be undertaken
between the European context described here and that of the United States or Canada. In
addition, further research could be undertaken with biopharming companies as well as
publicly funded research. The work of Einsiedel and Medlock (2005) on Canadian public
attitudes to biopharming may provide a useful starting point for developing such a
comparative study.

A second set of concerns focuses on the very use of biopharming as a case study. As
Beaulieu et al. (2007) point out, case studies are commonly the way contributions to the
development of science and technology studies are made. Yet this can mean that research
becomes little more than elaborately complicated descriptive reports which cannot be
extended beyond their own case and context - what Castree (2005) describes as an
‗epistemology of particulars‘. Similarly within STS Wyatt and Balmer (2007) have asked:

―Interesting as the story itself may be, how does it contribute to discussions of
anything beyond itself?‖ (Wyatt and Balmer 2007:1)

Wyatt and Balmer‘s comment comes in a call to rethink traditional ideas of ‗middle range‘
theories which can bridge the gap in STS between detailed and rich, but isolated, case
studies and grand theories of actor-networks or the social construction of technology. In
describing the ways in which biopharming is placed, I have suggested that it contributes the
elaboration of a 'geography of expectations'. The sociology of expectations has been
described (Geels 2007) as providing one means of bridging the gap described by Wyatt and
Balmer. Analysing the operation of futures and visions provides a means of examining how
the relations are established between actors to form the ‗assemblages‘, ‗networks‘ or
‗ensembles‘ of these models. Similarly, work which describes the role of place in shaping
and constituting relations between actors offers the potential to compare and contrast
between case studies. Together, they may allow an empirically rich and theoretically

289
productive approach to the examining the material, narrative and spatial patternings of
technoscience.

7.4 Future Directions

Having spent much of this thesis describing the construction of promising futures, it is now
time to introduce some hopeful visions of my own, suggesting forward directions for the
development of the current research. Before concluding, I suggest avenues through which
the conclusions of my thesis could be productively pursued.

The first of these is through a reconsideration of the relationships between the publics of
pharmaceutical and foods. As described above, different roles are available to lay actors in
these situations. While much STS research has focussed on the contribution of 'expert
patients' in the co-production of biomedical knowledge, other trends suggest the need to
expand social science analysis of biomedical publics. In particular, increased policy
emphasis on 'consumer healthcare' suggests the need to consider the opportunity for, and
ability of, publics to participate meaningfully in consumer-based medicine (Williams et al.
2008; Oudshoorn 2008; Webster et al. 2009; Morris, Armstrong, and Balmer 2009). Such
questions are accentuated in light of the rise of 'direct-to-consumer' products in previously
state-controlled health systems such as the NHS. The increasing availability of both
pharmaceuticals and predictive genetic testing through the internet may provide a
productive focus for such research.

Secondly, my discussion of containment and of the geographies of pharmaceutical


biotechnology may be usefully developed through engagement with work on biosecurity.
An increasingly large body of research has focussed on the means through which control is
asserted over the materialities and biologies of biotechnology, of invasive species and of
disease, as well as of biological weapons agents (Collier, Lakoff, and Rabinow 2004;
Donaldson and Wood 2004; Cooper 2006; Braun 2007; Hinchliffe and Bingham 2008;
Barker 2008; Rappert, Balmer, and Stone 2008). Much of this research concentrates on the
role of spatial, technological and political strategies in controlling unruly biologies to the

290
molecular level. Bringing discussion of the controlled, surveilled facilities of good
manufacturing practice together with this work may suggest containment's place in wider
practices of biosecurity, and will contribute to developing these studies.

Finally, the development of biopharming highlights the importance of 'imagined


geographies' in technological futures. These geographies establish a relationship between
innovators and users, and are performative in that their influence on technological
development makes these geographies material. The importance of the geographies of
expectations is likely to be seen beyond the individual case study of biopharming, not only
in technologies which target the developing world, but in those which posit new modes of
transport or energy production in combating global climate change.

7.5 Placing the Promise of Biopharming

To conclude, I return to the title of my thesis: 'No Natural Home'. Taken from the EMBO
paper which accompanied the launch of the Pharma-Planta Consortium (Ma et al. 2005), it
describes biopharming as a technology without an obvious place or nature. I have argued
that the places and natures of biopharming are constructed in relation to agriculture and
pharmaceuticals, for experts and publics. It is through this process of 'placing' that the
promise of biopharming is established and challenged, and through which its future
development will occur.

Whichever way this placing is narrated, the story of the thesis is one of the reshaping of the
promise of biopharming as it encounters and attempts to reconfigure relationships between
the publics, materials, places and regulations of agriculture and medicine. In equating the
standards of Western biopharmaceutical manufacture, and maintaining a separation
between foods and pharmaceuticals when they can no longer be easily distinguished
visually, the scale and natures that underpin the agricultural promise of biopharming are
lost. Instead, new places of agriculture and medicine are constructed, new relationships
between foods and drugs are formed and new promises emerge.

291
Looking beyond biopharming, I have described how the credibility of visions is constituted
through their relation to existing narratives of technological development - to the 'authority
of a parallel venture' (Said 1975) - and to the materials, natures and geographies of novel
and everyday technoscience. As a substantive contribution to the field of science and
technology studies, my research draws attention to the geographies of emerging
technoscience, and to the ways in which technoscientific imaginings take their place in the
world.

292
APPENDIX I
The recruitment letter

Department of Science and Technology Studies


University College London
Gower St.
London
WC1E 6BT
Dear ______,

I am writing to ask if the members of name of group would be able to help me with my
research. I am studying for a PhD at University College London, and am interested in how
people have been affected by changes in food and medicine over recent years, and how they
feel about some possible future changes.

I'm holding a series of focus groups around the country and was hoping your members
might be interested in forming one of these in location. I am looking for around 8 people
for a group which will meet twice rough date, for an hour and a half each time.

For their help with my research, I will give all those who participate £30.

I will also send a copy of this letter by email, and more information is available at
www.foodmedicinegenetics.org27, as well as in the attached fliers.

I can be contacted on 07870699208 at any time if you have any questions.

Best wishes,

Richard Milne

27
Archived at http://www.homepages.ucl.ac.uk/~ucrhrjm/research/mf.html
293
The group flyer

294
The information booklet provided to groups

295
296
297
298
299
APPENDIX II
Members of the Pharma-Planta Consortium

Country Institution

Austria University of Agricultural Sciences

BOKU (University of Natural Resources and Applied Sciences)

Polymun Scientific Immunbiologische Forschungs GmbH

Belgium Universite Catholique de Louvain

Vlaams Interuniversitair Instituut voor Biotechnologie (VIB)

France CIRAD (Centre de coopération internationale en recherche agronomique pour le


développement)

Universite Blaise Pascal Clermont-Ferrand II

Institut National de la Recherche Agronomique (INRA)

Germany Fraunhofer Aachen (2 groups)

RWTH Aachen (2 groups)

Ruprecht-Karls-Universität Heidelberg

Max Planck Institute, Postdam

Institut für Pflanzengenetik und Kulturpflanzenforschung (IPK), Gatersleben

Sartorius-Stedim Biotech, Goettingen

Greece Agricultural University of Athens

Ireland Maynooth University (2 groups)

Trinity College, Dublin

Italy University of Verona

ENEA (National Agency for New Technologies, Energy and the Environment)

Consiglio Nazionale Delle Ricerche, Rome, Italy

Netherlands Mosaic Systems BV, Prinsenbeek

300
Spain Universitat de Lleida

South Africa Council for Scientific and Industrial Research (CSIR), Pretoria

Sweden Diamyd Medical AB

Switzerland Universite de Neuchatel, Neuchatel,

UK St. George‘s Hospital Medical School (2 groups)

John Innes Centre (2 groups)

University of Cambridge (2 groups)

MIHR (Centre for Management of Intellectual Property in Health Care and


Development), Oxford

Oxford Brookes University

Rothamsted Research

University of Strathclyde

University of Leeds

University of Warwick

301
APPENDIX III

302
303
304
305
Prompts Presented to the Group in Meeting Two

―250 acres of greenhouse space would be sufficient to grow enough transgenic potato
plants to meet South East Asia‘s annual demand for the hepatitis B virus vaccine‖

(European Molecular Biology Organization Report written by members of the EU funded


Pharma-Planta Consortium, 2005)

―Since ancient times plants have served as a natural source of treatments and therapies such
as aspirin, quinine, and coffee‖

(Biotechnology Industry Organization, USA)

―these plants could cause irreversible harm to the natural environment, with potentially
devastating consequences for food production and biodiversity‖

(Greenpeace UK)

―PMP technology might be promising, offering possibilities for reduced cost, adequate
supply and effective human protein-based medicines‖

(International Association of Patients Organizations Report, 2005)

MOLECULAR FARMING IS AN AGRICULTURAL TECHNOLOGY

MOLECULAR FARMING IS A MEDICAL TECHNOLOGY

306
APPENDIX IV
The coding framework: themes and codes

Actors o money and


- Responsibility pharmaceuticals
- Prior behaviour of actors o need for research
- who makes food o Pharmaceutical good
- who does research intentions
o researchers and o GM good intentions
scientists o Goal of MF
  identities
 sources of info Identities
- Trust in practitioner - consumption
o Medical o individual choice
o Herbal o control
- Public  try to avoid
o public acceptance medicines
o  identities  try to avoid
- Pharmaceutical companies GM
- Protesters o trust in food
- Biopharming medical or o frequency of
agricultural by practitioner consumption
- Government o testing and
o Independence knowledge
o actors
Applications of Biopharming - Patients
- rabies - Personal relevance
- vaccines - Link with national identity 
- tobacco Places
- target choice
- HIV/AIDS Medical Imaginaries
- edible vaccines - medicines worthwhile
- medicines well regulated 
Futures Testing
- Promise - need for products
- Novelty - money and pharmaceuticals
- Prediction o pharmaceutical
- Expectations companies
o Patient expectations
- Motives Nature
o money and food - natural agency

307
- natural or not - Containment: Efficacy
o natural medicine o containment place
 medicines specific
because not o containment
natural ineffective
o natural food o containment
- ‗messing with nature‘ advantages
- illness natural - Containment: Types
o containment by
Pasts sterility
- ‗way we live now‘ o containment by plant
- industrialisation of food characteristics
o GM and industrial food o containment by plant
production body
o BSE o containment by
- tradition isolation
- Terminator tech o containment by
- loss of faith distance
- progress or inevitable o Greenhouses
- history or heritage  greenhouses
- precedents gone wrong small
- past success and vaccination  greenhouses
- industrialisation of food expensive
- changing views
- changing food advice ‘Placing Biopharming’
- changes in medicines - relation with other biotech
- changes in food technology - relation with medicines
- changes eating habits - relation with GM
o terminator tech
Places o  Pasts
- Consumption - relation with food
o where we eat - relation with farming
o where food bought - stem cells
- Production
o where food from Products
o where food grown - Biopharming medical or
o Fields agricultural by product
- Nations - Medicines
o regulatory differences o unintended
between countries consequences
o Europe  side effects
- Developing World allergies 
- scale of biopharming Testing
- Inside or outside o what is in medicine
- Field trials  types of
308
medicines
Production
- Foods - processed food
o 'bad' food/‘good‘ food - self-reported ignorance about
o what added to food food production
o unintended - chemicals
consequences - GMP  Places
o frequency of
consumption Risk
o processed food/fresh - risk to wildlife
food - risk to humans
  Production
o Choice Testing and Knowledge
 choice food: - unfamiliar
tradition - need to test
 choice food: - not ready
taste - too much information
 choice food: - Types of testing
quick o trial and error
 choice food: o scientific knowledge
quality uncertain
 choice food: o clinical trials
production o common sense and
 choice food: moderation
healthy - quality of information
 choice food: - sources of info
cost - unknowns
 choice food: - uncertainty
appearance - GM testing
- Regulation
- Product effect Trust
o how know its working - trust in regulation
- Product characteristics - trust in practitioner
o Feel of food - trust in information
o Glycosylation - trust in food
- Equivalence - trust in medicine

309
BIBLIOGRAPHY
Abraham, J., and G. Lewis. 2002. ―Citizenship, Medical Expertise and the Capitalist
Regulatory State in Europe.‖ Sociology 36:67―88.

Akrich, M. 1992. ―The description of technological objects.‖ Pp. 205-224 in Shaping


Technology/Building Society: Studies in Sociotechnical Change, edited by W. E.
Bijker and J. Law. Cambridge, MA: MIT Press.

Ammann, K. 2004. ―The role of science and discourse in the application of the
precautionary approach.‖ Pp. 291-302 in Molecular Farming: Plant-made
Pharmaceuticals and Technical Proteins, edited by R. Fischer and S. Schillberg.
Wiley VCH.

Anderson, B. 1983. Imagined Communities: Reflections on the Origin and Spread of


Nationalism. Verso.

Anderson, B., M. Kearnes, and R. Doubleday. 2007. ―Geographies of nano-technoscience.‖


Area 39:139―142.

Applbaum, K. 2006. ―Pharmaceutical Marketing and the Invention of the Medical


Consumer.‖ PLoS Medicine 3:e189.

Armstrong, V. et al. 2006. Public Perspectives on the Governance of Biomedical Research:


A Qualitative Study in a Deliberative Context. Wellcome Trust.

Arntzen, C., S. Plotkin, and B. Dodet. 2005. ―Plant-derived vaccines and antibodies:
potential and limitations.‖ Vaccine 23:1753-1756.

Atkinson, P. 2005. ―Qualitative Research—Unity and Diversity.‖ Forum: Qualitative


Social Research 6. http://www.qualitative-
research.net/index.php/fqs/article/viewArticle/4/9 (Accessed January 25, 2009).

Babusiaux, C., JY Le Deaut, D. Sicard, and J. Testard. 2003. ―Rapport à la suite du débat
sur les OGM et les essais au champ.‖ Paris, MATE.

Barker, K. 2008. ―Flexible boundaries in biosecurity: accommodating gorse in Aotearoa


New Zealand.‖ Environment and Planning A 40:1598-1614.

Barrett, K., and E. Abergel. 2000. ―Breeding familiarity: environmental risk assessment for
genetically engineered crops in Canada.‖ 2―12.

310
Barta, A. et al. 1986. ―The expression of a nopaline synthase - human growth hormone
chimaeric gene in transformed tobacco and sunflower callus tissue.‖ Plant
Molecular Biology 6:347-357.

Basaran, P., and E. Rodríguez-Cerezo. 2008. ―Plant Molecular Farming: Opportunities and
Challenges.‖ Critical Reviews in Biotechnology 28:153-172.

Bauer, M. 2002. ―Controversial medical and agri-food biotechnology: a cultivation


analysis.‖ Public Understanding of Science 11:111, 93.

Bauer, M., and G. Gaskell. 2002. ―Researching the public sphere of biotechnology.‖ Pp. 1-
21 in Biotechnology: The Making of a Global Controversy, edited by M. Bauer and
G. Gaskell. Cambridge University Press.

Bauer, M., and J. Gutterling. 2007. ―Issue Salience and Media Framing over 30 Years.‖ Pp.
113-130 in Genomics and Society: Legal, Ethical and Social Dimensions, edited by
George Gaskell and M. Bauer. Earthscan Publications Ltd.

BBC. 2004. ―Bayer deals blow to UK GM crops.‖ BBC, March 31


http://news.bbc.co.uk/1/hi/sci/tech/3584763.stm (Accessed May 18, 2009).

Beaulieu, A., A. Scharnhorst, and P. Wouters. 2007. ―Not Another Case Study: A Middle-
Range Interrogation of Ethnographic Case Studies in the Exploration of E-science.‖
Science Technology Human Values 32:672-692.

Beckwith, J. A., T. Hadlock, and H. Suffron. 2003. ―Public perceptions of plant


biotechnology--a focus group study.‖ New Genetics & Society 22:125-141.

Bedford, T., and J. Burgess. 2001. ―The focus-group experience.‖ Pp. 121–135 in
Qualitative methodologies for geographers: issues and debates, edited by M. Limb
and C. Dwyer. London: Arnold.

Bhattacharya, S. 2004. ―UK scientists to clone human embryos.‖ New Scientist, August 11.

Bijker, W. E. 1995. ―Sociohistorical Technology Studies.‖ Pp. 229-256 in Handbook of


Science and Technology Studies, edited by S. Jasanoff, G. Markle, J. Peterson, and
T. Pinch. London: Sage.

Bingham, N. 2008. ―Slowing things down: Lessons from the GM controversy.‖ Geoforum
39:111-122.

BIO. 2008. ―Guide to Biotechnology 2008.‖ http://bio.org/speeches/pubs/er/ (Accessed


May 8, 2009).

311
BIO. 2006. Industry Update on Plant-Made Pharmaceuticals. Biotechnology Industry
Association http://bio.org/healthcare/pmp/FruitionFactSheetDecember2006.pdf
(Accessed January 7, 2008).

Bischoff, F. 2004. ―A Top-down View of Molecular Farming from the Pharmaceutical


Industry: Requirements and Expectations.‖ Pp. 267-290 in Molecular Farming:
Plant-made Pharmaceuticals and Technical Proteins, edited by R. Fischer and S.
Schillberg. Wiley VCH.

Bonneuil, C., P.B. Joly, and C. Marris. 2008. ―Disentrenching Experiment: The
Construction of GM―Crop Field Trials As a Social Problem.‖ Science Technology
Human Values 33:201―229.

Borup, M., N. Brown, K. Konrad, and H. Van Lente. 2006. ―The sociology of expectations
in science and technology.‖ Technology Analysis & Strategic Management 18:285-
298.

Bowker, G. 2007. The Postcolonial and the Global. Univ Of Minnesota Press.

Bowker, G., and S.L. Star. 2000. Sorting Things Out: Classification and Its Consequences.

Bowring, F. 2002. Science, Seeds and Cyborgs: Biotechnology and the Appropriation of
Life. Verso Books.

Brandle, J. 2004. ―The Field Evaluation of Transgenic Crops Engineered to Produce


Recombinant Proteins.‖ Pp. 69-76 in Molecular Farming: Plant-made
Pharmaceuticals and Technical Proteins, edited by R. Fischer and S. Schillberg.
Wiley VCH.

Braun, B. 2007. ―Biopolitics and the molecularization of life.‖ Cultural Geographies 14: 6-
28

Brooks, S. 2005. ―Biotechnology and the Politics of Truth: From the Green Revolution to
an Evergreen Revolution.‖ Sociologia Ruralis 45:360―379.

Brown, N. 1998. ―Ordering Hope.‖ Unpublished PhD Thesis, School of Independent


Studies, Lancaster University.

Brown, N. 2000. ―Organising/Disorganising the Breakthrough Motif: Dolly the Cloned


Ewe Meets Astrid the Hybrid Pig.‖ Pp. 87-110 in Contested Futures: A Sociology of
Prospective Techno-science.

Brown, N. 2003. ―Hope Against Hype - Accountability in Biopasts, Presents and Futures.‖
Science Studies 16:3―21.

312
Brown, N. 2007. ―Shifting Tenses: Reconnecting Regimes of Truth and Hope.‖
Configurations 13:331–355.

Brown, N., and M. Michael. 2001. ―Switching between Science and Culture in Transpecies
Transplantation.‖ Science Technology Human Values 26:3―22.

Brown, N., and M. Michael. 2003. ―A Sociology of Expectations: Retrospecting Prospects


and Prospecting Retrospects.‖ Technology Analysis and Strategic Managment
15:18, 3.

Brown, N., and M. Michael. 2004. ―Risky creatures: institutional species boundary change
in biotechnology regulation.‖ Health, Risk & Society 6:207―222.

Brown, N., B. Rappert, and A. Webster. 2000. ―Introducing Contested Futures: From
Looking into the Future to Looking at the
Future.‖ Pp. 3-21 in Contested Futures: A Sociology of Prospective Techno-science,
edited by N. Brown, B. Rappert, and A. Webster. London: Ashgate.

Brown, N., and A. Webster. 2004. New medical technologies and society. Polity.

Bryman, A. 2001. Social Research Methods. Oxford University Press.

Bucchi, M., and F. Neresini. 2008. ―Science and public participation.‖ Pp. 449-473 in New
Handbook of Science and Technology Studies, edited by E. Hackett, O.
Amsterdamska, M. Lynch, and J. Wajcman. MIT Press.

Bud, R. 1991. ―Biotechnology in the Twentieth Century.‖ Social Studies of Science 21:415-
457.

Bud, R. 1993. The Uses of Life: A History of Biotechnology. Cambridge University Press.

Burchell, K. 2007. ―Boundary Work, Associative Argumentation and Switching in the


Advocacy of Agricultural Biotechnology.‖ Science as Culture 70, 49.

Burgess, J. 1996. ―Focusing on Fear: The Use of Focus Groups in a Project for the
Community Forest Unit, Countryside Commission.‖ Area 28:130-135.

Burgess, J., M. Limb, and C. M. Harrison. 1988a. ―Exploring environmental values through
the medium of small groups: 1. Theory and practice.‖ Environment and Planning A
20:309-326.

Burgess, J., M. Limb, and C. M. Harrison. 1988b. ―Exploring environmental values through
the medium of small groups: 2. Illustrations of a group at work.‖ Environment and
Planning A 20:257-276.
313
Burgess, J. et al. 2007. ―Deliberative mapping: a novel analytic-deliberative methodology
to support contested science-policy decisions.‖ Public Understanding of Science
16:299―322.

Burke, D. 2004. ―GM food and crops: what went wrong in the UK?.‖ EMBO Reports
5:432-436.

Burke, D. 1998. ―The `yuk' factor.‖ in Consuming Passions: Cooking and Eating in the Age
of Anxiety, edited by Sian Griffiths and Jennifer Wallace. Mandolin.

Busby, H. & Martin, P., 2006. ―Biobanks, national identity and imagined communities: The
case of UK Biobank‖. Science as Culture, 15, 237-251.

Bush, V. 1945. Science the Endless Frontier. Washington: Office of Scientific Research
and Development http://www.nsf.gov/od/lpa/nsf50/vbush1945.htm (Accessed
January 5, 2009).

Byrne, B., 2004. Qualitative Interviewing. In C. Seale, ed. Researching Society and
Culture. London: Sage Publications Inc, pp. 180-191.

Callon, M. 1986. ―Some Elements of a Sociology of Translation: Domestication of the


Scallops and the Fishermen.‖ Power, Action and Belief: A New Sociology of
Knowledge 196-223.

Calnan, M., D. Montaner, and R. Horne. 2005. ―How acceptable are innovative health-care
technologies? A survey of public beliefs and attitudes in England and Wales.‖
Social Science & Medicine 60:1937―1948.

Calnan, M., and S. Williams. 1992. ―Images of scientific medicine..‖ Sociology of Health &
Illness 14:233―254.

Carey, M. 1995. ―Comment: Concerns in the Analysis of Focus Group Data.‖ Qual Health
Res 5:487―495.

Castree, N. 2005. ―The epistemology of particulars: Human geography, case studies and
'context'.‖ Geoforum 36:541-544.

Cohen, D., M. McCubbin, J. Collin, and G. Perodeau. 2001. ―Medications as Social


Phenomena.‖ Health (London) 5:441-469.

Collier, S., A. Lakoff, and P. Rabinow. 2004. ―Biosecurity: Towards an anthropology of the
contemporary.‖ Anthropology Today 3―7.

314
Commandeur, U., and R. M Twyman. 2004. ―Biosafety Aspects of Molecular Farming in
Plants.‖ Pp. 251-266 in Molecular Farming: Plant-made Pharmaceuticals and
Technical Proteins, edited by R. Fischer and S. Schillberg. Wiley VCH.

Commandeur, U., R. M. Twyman, and R. Fischer. 2003. ―The biosafety of molecular


farming in plants.‖ AgBiotechNet [Online] 5:110.

Constable, A. et al. 2007. ―History of safe use as applied to the safety assessment of novel
foods and foods derived from genetically modified organisms.‖ Food and Chemical
Toxicology 45:2513-2525.

Conway, G. 1999. The Doubly Green Revolution: Food for All in the Twenty-First Century.
Cornell University Press.

Cook, I., and M. Crang. 1995. Doing ethnographies. University of East Anglia.

Cooper, M. 2006. ―Pre-empting Emergence: The Biological Turn in the War on Terror.‖
Theory Culture Society 23:113―135.

Coyle, F., and J. Fairweather. 2005. ―Space, time and nature: exploring the public reception
of biotechnology in New Zealand.‖ Public Understanding of Science 14:143―161.

Cunningham-Burley, S., A. Kerr, and S. Pavis. 1999. ―Theorizing subjects and subject
matter in focus group research‖ edited by J. Kitzinger and R. Barbour. Developing
focus group research: Politics, theory and practice 186-199.

CWR&D. 2005. ―Public engagement exercise on non-food agriculture.‖


http://www.aebc.gov.uk/aebc/subgroups/non_food_agriculture.shtml.

Dahl, R. 1967. Charlie and the Chocolate Factory. George Allen and Unwin.

Daniell, H., O. Carmona-Sanchez, and B. Burns. 2004. ―Chloroplast Derived Antibodies,


Biopharmaceuticals and Edible Vaccines.‖ Pp. 113-134 in Molecular Farming:
Plant-made Pharmaceuticals and Technical Proteins, edited by R. Fischer and S.
Schillberg.

Davies, G. 2006. ―The sacred and the profane: biotechnology, rationality, and public
debate.‖ Environment and Planning A 38:423 – 443.

Davies, G., and C. Dwyer. 2008. ―Progress reports.‖ Progress in Human Geography
32:399-406.

Davies, G., and C. Dwyer. 2007. ―Qualitative methods: are you enchanted or are you
alienated?.‖ Progress in Human Geography 31:257-266.

315
Department of Health. 2004. ―Choosing Health: Making healthy choices easier.‖
http://www.dh.gov.uk/en/Publicationsandstatistics/Publications/PublicationsPolicy
AndGuidance/DH_4094550 (Accessed April 19, 2009).

Desmond, M. 2004. ―Methodological challenges posed in studying an elite in the field.‖


Area 36:262―269.

Deuten, J. J., and A. Rip. 2000. ―The narrative shaping of a product creation process.‖
Brown et al.(2000) 65–86.

Donaldson, A., and D. Wood. 2007. ―Avian influenza and events in political
biogeography.‖ Area 39:128-130.

Donaldson, A., and D. Wood. 2004. ―Surveilling strange materialities: categorisation in the
evolving geographies of FMD biosecurity.‖ Environment and Planning D: Society
and Space 22:373―391.

Doubleday, R. 2004. ―Institutionalising non-governmental organisation dialogue at


Unilever: framing the public as consumer-citizens.‖ Science and Public Policy
31:117-126.

Douglas, M. 2002. Purity and Danger: An Analysis of the Concepts of Pollution and
Taboo. 1st ed. Routledge.

DTI. 2003. GM Nation? The findings of the public debate. London: Department of Trade
and Industry.

Dunwell, J. M. 2005. ―Technologies for biological containment of GM and non-GM crops.


Defra contract CPEC 47. Department for Environment.‖ Food and Rural Affairs
(Defra), London.

Durant, J, G Evans, and G Thomas. 1992. ―Public understanding of science in Britain: the
role of medicine in the popular representation of science.‖ Public Understanding of
Science 1:161-182.

Eames, M., W. Mcdowall, M. Hodson, and S. Marvin. 2006. ―Negotiating contested visions
and place-specific expectations of the hydrogen economy.‖ Technology Analysis &
Strategic Management 18:361-374.

EC. 2003. ―Commission Directive 2003/94/EC laying down the principles and guidelines
of good manufacturing practice in respect of medicinal products for human use and
investigational medicinal products for human use.‖.

316
EC. 2008. ―EudraLex The Rules Governing Medicinal Products in the European Union:
Volume 4
EU Guidelines to Good Manufacturing Practice.‖.

Eden, S., C. Bear, and G. Walker. 2008. ―Mucky carrots and other proxies: Problematising
the knowledge-fix for sustainable and ethical consumption.‖ Geoforum 39:1044-
1057.

EFPIA. 2008. The Pharmaceutical Industry in Figures. http://www.efpia.org/ (Accessed


January 6, 2009).

EFSA. 2006. ―Guidance Document of the Scientific Panel on Genetically Modified


Organisms for the Risk Assessment of Genetically Modified Plants and Derived
Food and Feed.‖.

EFSA. 2008. ―Draft Opinion of the Scientific Panel on Genetically Modified


Organisms on the risk assessment of genetically modified plants
used for non-food or non-feed purposes.‖.

Einsiedel, E. F., and J. Medlock. 2005. ―A public consultation on plant molecular farming.‖
AgBioForum 8:26-32.

Ely, A. 2008. ―Safety is just the start if we want good regulation.‖ Food Ethics 3:6-8.

EMEA. 2002. ―CPMP/BWP/764/02 Points to Consider on Quality Aspects of Medicinal


Products Containing Active Substances Produced by Stable Transgene Expression
in Higher Plants.‖.

EMEA. 2006. ―EMEA/CHMP/BWP/48316/2006 Guideline on the quality of biological


active substances produced by stable transgene expression in higher plants [Draft
2].‖.

EMEA. 2008. ―EMEA/CHMP/BWP/48316/2006 Guideline on the quality of biological


active substances produced by stable transgene expression in higher plants [Final].‖.

EU. 2002. ―CORDIS FP6: What is FP6.‖ http://cordis.europa.eu/fp6/whatisfp6.htm


(Accessed March 7, 2007).

EU. 2009. Agriculture in the European Union - Statistical and economic information 2008.
http://ec.europa.eu/agriculture/agrista/index_en.htm.

Faye, L., A. Boulaflous, M. Benchabane, V. Gomord, and D. Michaud. 2005. ―Protein


modifications in the plant secretory pathway: current status and practical
implications in molecular pharming..‖ Vaccine 23:1770―1778.

317
Feagan, R. 2007. ―The place of food: mapping out the 'local' in local food systems.‖
Progress in Human Geography 31:23-42.

Felt, U. et al. 2007. Taking European Knowledge Society Seriously: Report of the Expert
Group on Science and Governance to the Science, Economy and Society
Directorate, Directorate-General for Research, European Commission. Directorate-
General for Research, European Commission.

Finnegan, D. 2008. ―The Spatial Turn: Geographical Approaches in the History of


Science.‖ Journal of the History of Biology 41:369-388.

Fischer, R., and N. Emans. 2000. ―Molecular farming of pharmaceutical proteins.‖


Transgenic Research 9:279-299.

Fischer, R., and S. Schillberg, eds. 2004. Molecular Farming: Plant-made Pharmaceuticals
and Technical Proteins. Wiley VCH.

Fox, J. 2003. ―Puzzling industry response to ProdiGene fiasco.‖ Nat Biotech 21:3-4.

Frankland, J., and M. Bloor. 1999. ―Some issues arising in the systematic analysis of focus
group materials.‖ Pp. 144-155 in Developing Focus Group Research: Politics,
Theory and Practice, edited by J. Kitzinger and R. Barbour. Sage Publications Ltd.

Franklin, S. 2001. ―Culturing biology: cell lines for the second millennium.‖ Health 5:335-
354.

Franklin, S. 2006. ―The Cyborg Embryo: Our Path to Transbiology.‖ Theory Culture
Society 23:167-187.

Frewer, L., and B. Salter. 2002. ―Public attitudes, scientific advice and the politics of
regulatory policy: the case of BSE.‖ Science and Public Policy 29:137-145.

Galambos, L., and J. L. Sturchio. 1998. ―Pharmaceutical firms and the transition to
biotechnology: A study in strategic innovation.‖ The Business History Review 250-
278.

Gaskell, G. 2004. ―Science policy and society: the British debate over GM agriculture.‖
Current Opinion in Biotechnology 15:245, 241.

Gaskell, G., and M. Bauer, eds. 2007. Genomics and Society: Legal, Ethical and Social
Dimensions. Earthscan Publications Ltd.

Gaskell, G. et al. 2006. Europeans and Biotechnology in 2005. Patterns and Trends. Final
Report on Eurobarometer 64.3. Brussels: European Commission‘s Directorate-

318
General for Research< http://www. ec. europa. eu/research/press/2006/pr1906en.
cfm.

GE3LS Alberta. 2007. Pharming the Future: A Citizens' Panel Perspective on Plant
Molecular Farming. http://www.fp.ucalgary.ca/pharmingthefuture/.

Geels, F. 2007. ―Feelings of Discontent and the Promise of Middle Range Theory for STS:
Examples from Technology Dynamics.‖ Science Technology Human Values
32:627-651.

Geels, F., and W. Smit. 2000. ―Failed technology futures: pitfalls and lessons from a
historical survey.‖ Futures 32:867―885.

Geesink, I., B. Prainsack, and S. Franklin. 2008. ―Stem Cell Stories 1998–2008.‖ Science
as Culture 17:1-11.

Gibbs, A. 1997. Focus Groups. University of Surrey


http://sru.soc.surrey.ac.uk/SRU19.html.

Gieryn, T. 1983. ―Boundary-Work and the Demarcation of Science from Non-Science:


Strains and Interests in Professional Ideologies of Scientists.‖ American
Sociological Review 48:795, 781.

Gieryn, T. 2002. ―What Buildings Do.‖ Theory and Society 31:35―74.

Goffman, E. 1974. Frame Analysis: An Essay on the Organization of Experience.


Northeastern University Press.

Gomord, V., and L. Faye. 2004. ―Posttranslational modification of therapeutic proteins in


plants.‖ Current opinion in plant biology 7:171-181.

Gomord, V. et al. 2004. ―Production and glycosylation of plant-made pharmaceuticals: the


antibodies as a challenge.‖ Plant Biotechnology Journal 2:83-100.

Gomord, V., P. Chamberlain, R. Jefferis, and L. Faye. 2005. ―Biopharmaceutical


production in plants: problems, solutions and opportunities.‖ Trends in
Biotechnology 23:559-565.

Good, M.J. 2001. ―The Biotechnical Embrace.‖ Culture, Medicine and Psychiatry 25:395-
410.

Goodman, D. 1999. ―Agro-Food Studies in the'Age of Ecology': Nature, Corporeality, Bio-


Politics.‖ Sociologia Ruralis 39:17-38.

319
Goodman, D. 2001. ―Ontology Matters: The Relational Materiality of Nature and Agro-
Food Studies.‖ Sociologia Ruralis 41:182-200.

Goodyear, M.D.E. 2006. ―Further lessons from the TGN1412 tragedy.‖ BMJ 270-271.

Gottweis, H. 1998. Governing Molecules: The Discursive Politics of Genetic Engineering


in Europe and the United States. MIT Press.

Green, J., A. Draper, and E. Dowler. 2003. ―Short cuts to safety: risk and 'rules of thumb' in
accounts of food choice.‖ Health, Risk & Society 5:33―52.

Grunert, K. G. 2002. ―Current issues in the understanding of consumer food choice.‖


Trends in Food Science & Technology 13:275-285.

Hague, E. 2004. ―Key Thinkers on Space and Place.‖ edited by P. Hubbard, R. Kitchin, and
G. Valentine. Sage Publications Ltd.

Hall, S. 2002. Invisible Frontiers: The Race to Synthesize a Human Gene. New Ed. Oxford
University Press Inc, USA.

Hammersley, M., and P. Atkinson. 1994. Ethnography: Principles in Practice. 2nd ed.
Routledge.

Hayden, C. 2007. ―A Generic Solution? Pharmaceuticals and the Politics of the Similar in
Mexico.‖ Current Anthropology 48:475-495.

Hedgecoe, A., and P. Martin. 2003. ―The Drugs Don't Work: Expectations and the Shaping
of Pharmacogenetics.‖ Social Studies of Science 33:327-364.

Henke, C. 2000. ―Making a Place for Science: The Field Trial.‖ Social Studies of Science
30:483―511.

Henke, C., and T. Gieryn. 2008. ―Sites of Scientific Practice: The Enduring Importance of
Place,.‖ Pp. 353-377 in New Handbook of Science and Technology Studies, edited
by E. Hackett, O. Amsterdamska, M. Lynch, and J. Wajcman. MIT Press.

Hiatt, A., R. Cafferkey, and K. Bowdish. 1989. ―Production of antibodies in transgenic


plants.‖ Nature 342:76-78.

Hilgartner, S. 2006. ―Depicting Pasts, Projecting Futures: Making Histories of the New
Biology.‖ http://collab.nlm.nih.gov/webcastsandvideos/genomics/genomics.html
(Accessed September 30, 2008).

Hinchliffe, S., and N. Bingham. 2008. ―Securing life: the emerging practices of
biosecurity.‖ Environment and Planning A 40:1534-1551.
320
Holbrook, B., and P. Jackson. 1996. ―Shopping around: Focus Group Research in North
London.‖ Area 28:136-142.

Hopkins, M., P. Martin, P. Nightingale, A. Kraft, and S. Mahdi. 2007. ―The myth of the
biotech revolution: An assessment of technological, clinical and organisational
change.‖ Research Policy 36:566―589.

Hopkins, P. 2007. ―Thinking critically and creatively about focus groups.‖ Area 39:528-
535.

Horlick-Jones, T. et al. 2006. ―On evaluating the 'GM Nation?' Public debate about the
commercialisation of transgenic crops in Britain.‖ New Genetics and Society
25:265―288.

Horst, M. 2007. ―Public Expectations of Gene Therapy: Scientific Futures and Their
Performative Effects on Scientific Citizenship.‖ Science Technology Human Values
32:150―171.

Irwin, A. 2001. Sociology and the Environment: A Critical Introduction to Society, Nature,
and Knowledge. Polity Press.

Irwin, A., and M. Michael. 2003. Science, Social Theory and Public Knowledge. Open.

James, C. 2004. Global Status of Commercialized Biotech/GM Crops: 2004. Ithaca, NY:
ISAAA.

James, C. 2007. Global Status of Commercialized Biotech/GM Crops: 2007. Ithaca, NY:
ISAAA.

Jasanoff, S., 1995. ―Product, process, or programme: three cultures and the regulation of
biotechnology‖.Pp. 311-331 In M. Bauer, ed. Resistance to New Technology:
Nuclear Power, Information Technology and Biotechnology. Cambridge University
Press .

Jasanoff, S., 2004. Ordering knowledge, ordering society. In S. Jasanoff, ed. States of
Knowledge: The Co-Production of Science and Social Order. New York:
Routledge, pp. 13-45.

Jasanoff, S. 2005. Designs on Nature: Science and Democracy in Europe and the United
States. Princeton University Press.

Jasanoff, S. 2006. ―Biotechnology and Empire: The Global Power of Seeds and Science.‖
Osiris 21:273―292.

321
Jess, P., and D. Massey. 1995. ―The contestation of place.‖ A Place in the World 133-174.

Joly, P.B., and A. Kaufmann. 2008. ―Lost in Translation? The Need for 'Upstream
Engagement' with Nanotechnology on Trial.‖ Science as Culture 17:225―247.

Kamarck, M. 2006. ―Building biomanufacturing capacity- the chapter and verse.‖ Nat
Biotech 24:503-505.

Kearnes et al. 2006. ―From Bio to Nano: Learning Lessons from the UK Agricultural
Biotechnology Controversy.‖ Science as Culture 15:291―307.

Kerr, A., S. Cunningham-Burley, and A. Amos. 1997. ―The new genetics: professionals'
discursive boundaries.‖ The Sociological Review 45:279-303.

Kerr, A., S. Cunningham-Burley, and A. Amos. 1998. ―Drawing the line: an analysis of lay
people's discussions about the new genetics.‖ Public Understanding of Science
7:133, 113.

Khan, M., and L. Manderson. 1992. ―Focus groups in tropical diseases research.‖ Health
Policy Plan. 7:56―66.

Kirk, D., and K. Mcintosh. 2005. ―Social Acceptance of Plant-Made Vaccines: Indications
from a Public Survey.‖ AgBioForum 8. http://www.agbioforum.org/v8n4/v8n4a05-
kirk.htm.

Kitzinger, J. 1995. ―Qualitative Research: Introducing focus groups.‖ BMJ 311:299―302.

Kitzinger, J., and R. Barbour, eds. 1999. Developing Focus Group Research: Politics,
Theory and Practice. Sage Publications Ltd.

Kitzinger, J., and R. Barbour. 1999. ―Introduction: The Challenge and Promise of Focus
Groups.‖ Pp. 1-21 in Developing Focus Group Research: Politics, Theory and
Practice, edited by J. Kitzinger and R. Barbour. Sage Publications Ltd.

Kloppenburg, J. 2004. First the Seed: The Political Economy of Plant Biotechnology
(Science & Technology in Society). 2nd ed. University of Wisconsin Press.

Knight, A. J. 2006. ―Does Application Matter? An Examination of Public Perception of


Agricultural Biotechnology Applications.‖ AgBioForum 9.
http://www.agbioforum.org/v9n2/v9n2a07-knight.htm.

Knorr-Cetina, K. 1999. Epistemic Cultures: How the Sciences Make Knowledge. Harvard
University Press.

322
Koch, L. 2006. ―Past Futures: On the Conceptual History of Eugenics—a Social
Technology of the Past.‖ Technology Analysis & Strategic Management 18:329-
344.

Kohler, R. 2003. Landscapes and Labscapes: Exploring the Lab-field Border in Biology.
University of Chicago Press.

Kotchetkova, I., R. Evans, and S. Langer. 2008. ―Articulating Contextualized Knowledge:


Focus Groups and/as Public Participation?.‖ Science as Culture 17:71―84.

Krimsky, S. 2005. ―From Asilomar to industrial biotechnology: Risks, reductionism and


regulation.‖ Science as Culture 309―323.

Krohn, W., and J. Weyer. 1994. ―Society as a laboratory: the social risks of experimental
research.‖ Science and Public Policy 21:173-173.

Krumholz, H., J. Ross, A. Presler, and D. Egilman. 2007. ―What have we learnt from
Vioxx?.‖ BMJ 334:120-123.

Kusnadi, A. R., Z. L. Nikolov, and J. A. Howard. 2000. ―Production of recombinant


proteins in transgenic plants: Practical considerations.‖ Biotechnology and
Bioengineering 56:473-484.

Latour, B. 1983. ―Give Me a Laboratory and I will Raise the World.‖ P. 141―170 in
Science Observed, edited by K. Knorr and M. Mulkay. Sage http://www.bruno-
latour.fr/articles/article/12-GIVE%20ME%20A%20LAB.pdf.

Latour, B. 1988. Science in Action: How to Follow Scientists and Engineers Through
Society. Harvard University Press.

Latour, B. 1993. We Have Never Been Modern. Harvard University Press.

Latour, B. 2005. Reassembling the Social: An Introduction to Actor-network-theory


(Clarendon Lectures in Management Studies). Oxford University Press.

Law, J. 1992. ―Notes on the theory of the actor-network: Ordering, strategy, and
heterogeneity.‖ Systemic Practice and Action Research 5:379-393.

Law, J. 1994. Organizing Modernity: Social Order and Social Theory. WileyBlackwell.

Law, J. 2002. Aircraft Stories: Decentering the Object in Technoscience. Duke University
Press.

Law, J., and A. Mol. 2001. ―Situating technoscience: an inquiry into spatialities.‖
Environment and Planning D: Society and Space 19:609―621.
323
Lawrence, F. 2004. Not on the Label: What Really Goes into the Food on Your Plate.
Penguin.

Lemonick, M. 2002. ―Tomato Vaccine.‖ Time, November 18


http://www.time.com/time/magazine/article/0,9171,1003703,00.html (Accessed
May 11, 2009).

Levidow, L., and S. Carr. 2007. ―GM crops on trial: Technological development as a real-
world experiment.‖ Futures 39:408―431.

Levidow, L., J. Murphy, and S. Carr. 2007. ―Recasting "Substantial


Equivalence":Transatlantic Governance of GM Food.‖ Science Technology Human
Values 32:26―64.

Lezaun, J. 2006. ―Creating a New Object of Government: Making Genetically Modified


Organisms Traceable.‖ Social Studies of Science 36:499―531.

Lezaun, J. 2007. ―A market of opinions: the political epistemology of focus groups.‖ The
Sociological Review 55:130―151.

Lezaun, J., and Y. Millo. 2006. ―Regulatory experiments: genetically modified crops and
financial derivatives on trial.‖ http://eprints.lse.ac.uk/16034/ (Accessed December
20, 2008).

Livingstone, D. 2003. Putting Science in Its Place: Geographies of Scientific Knowledge.


Chicago and London: University Of Chicago Press.

Locating Technoscience. 2008. ―Locating the Future in and of Technoscience.‖ Locating


Technoscience Web Reader version 1.0. http://www.ucl.ac.uk/sts/locating-
technoscience/reader/?p=85 (Accessed November 6, 2008).

Longstaff, C., C. Whitton, R. Stebbings, and E. Gray. 2009. ―How do we assure the quality
of biological medicines?.‖ Drug Discovery Today 14:50-55.

Losey, J., L. Rayor, and M. Carter. 1999. ―Transgenic pollen harms monarch larvae.‖
Nature 399:214.

Ma, J.K. et al. 1995. ―Generation and assembly of secretory antibodies in plants.‖ Science
268:716-719.

Ma, J.K., P.M. Drake, and P. Christou. 2003. ―The production of recombinant
pharmaceutical proteins in plants.‖ Nat Rev Genet 4:794―805.

324
Ma, J.K., E. Barros, et al. 2005. ―Molecular farming for new drugs and vaccines..‖ EMBO
Rep 6:599, 593.

Ma, J.K., R. Chikwamba, et al. 2005. ―Plant-derived pharmaceuticals - the road forward.‖
Trends in Plant Science 10:585, 580.
Marcus, G. E. 1995. Technoscientific Imaginaries: Conversations, Profiles, and Memoirs.
University Of Chicago Press.

Marris, C., B. Wynne, P. Simmons, and S. Weldon. 2001. ―Public Perceptions of


Agricultural Biotechnology in Europe: Final Report of the PABE Research Project
Commission of European Communities, Contract No.‖ FAIR CT98-3844 (DG12-
SSMI).

Martin, P., N. Brown, and A. Kraft. 2008. ―From Bedside to Bench? Communities of
Promise, Translational Research and the Making of Blood Stem Cells.‖ Science as
Culture 17:29―41.

Martin, P., N. Brown, and A. Turner. 2008. ―Capitalizing hope: the commercial
development of umbilical cord blood stem cell banking.‖ New Genetics and Society
27:127-143.

Mason, H S, D M Lam, and C J Arntzen. 1992. ―Expression of hepatitis B surface antigen


in transgenic plants.‖ Proceedings of the National Academy of Sciences of the
United States of America 89:11745-11749.

Mason, H. S., H. Warzecha, T. Mor, and C. J. Arntzen. 2002. ―Edible plant vaccines:
applications for prophylactic and therapeutic molecular medicine.‖ Trends in
Molecular Medicine 8:324-329.

Massey, D., J. Allen, and P. Sarre. 1999. Human Geography Today. Polity Press.

Mayer, S. 2004. ―Comment: Is this the end for GM food?.‖ The Guardian, May 11.

Mayer, S., and A. Stirling. 2004. ―GM crops: good or bad?.‖ EMBO reports 5:1021.

McAfee, K. 2003a. ―Corn Culture and Dangerous DNA.‖ Journal of Latin American
Geography 2:18-42.

McAfee, K. 2003b. ―Neoliberalism on the molecular scale. Economic and genetic


reductionism in biotechnology battles.‖ Geoforum 34:203―219.

Merton, R., and E. Barber. 2004. The Travels and Adventures of Serendipity: A Study in
Sociological Semantics and the Sociology of Science. New edition. Princeton
University Press.

325
Michael, M. 1996. ―Ignoring science: discourses of ignorance in the public understanding
of science.‖ P. 107 in Misunderstanding Science?: The Public Reconstruction of
Science and Technology, edited by B. Wynne and A. Irwin. Cambridge: Cambridge
University Press.

Michael, M. 1998. ―Between citizen and consumer: Multiplying the meanings of the 'public
understanding of science'.‖ Public Understanding of Science 7:313―327.

Michael, M. 2000. Reconnecting Culture, Technology and Nature (International Library of


Sociology). Routledge.

Michael, M. 2000. ―Futures of the Present: From Performativity to Prehension.‖ Contested


Futures—A Sociology of Prospective Techno-Science. Aldershot: Ashgate 21-39.

Michael, M. 2002. ―Comprehension, Apprehension, Prehension: Heterogeneity and the


Public Understanding of Science.‖ Science, Technology, & Human Values
27:357―378.

Michael, M. 2006. Technoscience and Everyday Life: The Complex Simplicities of the
Mundane. Open University Press.

Michael, M., and N. Brown. 2004. ―The meat of the matter: grasping and judging
xenotransplantation.‖ Public Understanding of Science 13:397, 379.

Michael, M., and N. Brown. 2005a. ―Full Research Report. Xenotransplantation: Risk
Identities and the Human/Nonhuman Interface..‖ (Accessed January 11, 2008).

Michael, M., and N. Brown. 2005b. ―Scientific citizenships: self-representations of


xenotransplantation's publics.‖ Science as Culture 14:39―57.

Midgley, M. 2000. ―Biotechnology and monstrosity. Why we should pay attention to the
"yuk factor".‖ The Hastings Center Report 30:7-15.

Millstone, E., E. Brunner, and S. Mayer. 1999. ―Beyond `substantial equivalence'.‖ Nature
401:525-526.

Milne, R. 2009. ―Public Attitudes Toward Molecular Farming in the UK.‖ AgBioForum 11.
http://www.agbioforum.org/v11n2/v11n2a04-milne.htm (Accessed February 9,
2009).

Moreira, T., and P. Palladino. 2005. ―Between truth and hope: on Parkinson's disease,
neurotransplantation and the production of the'self'.‖ History of the Human Sciences
18:55.

326
Morgan, D. 1996. Focus Groups as Qualitative Research (Qualitative Research Methods).
Sage Publications, Inc.

Morris, N. 2003. ―Biological medicines in the age of biotech: public policy issues.‖
Regulation of the Pharmaceutical Industry 125.

Morris, N., V. Armstrong, and B. Balmer. 2009. ―Constructing a safe research


environment: Technology talk between researchers and volunteer research
subjects.‖ Health, Risk & Society 11:99-116.

Mulkay, M. 1993. ―Rhetorics of Hope and Fear in the Great Embryo Debate.‖ Social
Studies of Science 23:721―742.

Murdoch, J. 2005. Post-structuralist Geography: A Guide to Relational Space. Sage


Publications Ltd.

Murdoch, J., T. Marsden, and J. Banks. 2000. ―Quality, Nature, and Embeddedness: Some
Theoretical Considerations in the Context of the Food Sector.‖ Economic
Geography 76:107-125.

Myers, G. 2000. ―Displaying opinions: Topics and disagreement in focus groups.‖


Language in Society 27:85―111.

Myers, G. 1991. ―Stories and Styles in Two Molecular Biology Review Articles.‖ in
Textual Dynamics of the Professions: Historical and Contemporary Studies of
Writing in Professional Communities, edited by C. Bazerman and J. Paradis.
Madison, Wisconsin: University of Wisconsin Press.

Nagle, T., C. Berg, R. Nassr, and K. Pang. 2003. ―The further evolution of biotech.‖ Nat
Rev Drug Discov 2:75-79.

Naylor, S. 2005. ―Introduction: historical geographies of science; places, contexts,


cartographies.‖ The British Journal for the History of Science 38:1―12.

Nevitt, J., B. F Mills, D. W Reaves, and G. W Norton. 2003. ―Public Perceptions of


Tobacco Biopharming.‖ AgBioForum 9. http://www.agbioforum.org/v9n2/v9n2a05-
nevitt.htm.

Nightingale, P., and P. Martin. 2004. ―The myth of the biotech revolution.‖ Trends in
Biotechnology 22:564―569.

OED. 2008. Oxford English Dictionary. Oxford.

Ophir, A., and S. Shapin. 1991. ―The Place of Knowledge A Methodological Survey.‖
Science in Context 4:3-22.
327
Oudshoorn, N. 2008. ―Diagnosis at a distance: the invisible work of patients and healthcare
professionals in cardiac telemonitoring technology.‖ Sociology of Health & Illness
30:272-288.

Petryna, A. 2005. ―Ethical variability: Drug development and globalizing clinical trials.‖
American Ethnologist 32:183-197.

Pisano, G. P. 2002. ―Pharmaceutical biotechnology.‖ Pp. 347-366 in Technological


Innovation and Economic Performance, edited by B. Steil, D.G. Victor, and R.R.
Nelson. Princeton University Press.

Plein, C. 1991. ―Popularizing Biotechnology: The Influence of Issue Definition.‖ Science,


Technology, and Human Values 16:490, 474.

Plows, A., D. Wall, and B. Doherty. 2004. ―Covert repertoires: ecotage in the UK 1.‖
Social Movement Studies 3.

Pollan, M. 2007. The Omnivore's Dilemma: The Search for a Perfect Meal in a Fast-food
World. New edition. Bloomsbury Publishing PLC.

Pollock, A. M. 2004. NHS plc. Verso London.

Powell, R. 2007. ―Geographies of science: histories, localities, practices, futures.‖ Prog


Hum Geogr 31:309―329.

PPC. 2004. ―Pharma-Planta Press Release.‖ http://www.pharma-planta.org/newsarch.htm


(Accessed February 4, 2006).

PPC. 2005. ―Statement of Intent on Humanitarian Use.‖ http://www.pharma-


planta.org/resources_documents/SINTfinal.pdf.

PPC. 2007. ―Third Periodic Activity Report of the Pharma-Planta Consortium.‖.

Rabeharisoa, V., and M. Callon. 2004. ―Patient and Scientists in French Muscular
Dystrophy Research.‖ in States of Knowledge: The Co-production of Science and
Social Order, edited by S. Jasanoff. London: Routledge.

Rajan, K.S. 2006. Biocapital: The Constitution of Postgenomic Life. Duke University Press.

Ramessar, K., T. Capell, and P. Christou. 2008. ―Molecular pharming in cereal crops.‖
Phytochemistry Reviews 7:579-592.

328
Ramessar, K., T. Rademacher, et al. 2008. ―Cost-effective production of a vaginal protein
microbicide to prevent HIV transmission.‖ Proceedings of the National Academy of
Sciences 105:3727―3732.

Ramessar, K., M. Sabalza, T. Capell, and P. Christou. 2008. ―Maize plants: An ideal
production platform for effective and safe molecular pharming.‖ Plant Science
174:409―419.

Rappert, B., B. Balmer, and J. Stone. 2008. ―Science, Technology and the Military:
Priorities, Preoccupations, and Possibilities.‖ Pp. 719-741 in New Handbook of
Science and Technology Studies, edited by E. Hackett, O. Amsterdamska, M.
Lynch, and J. Wajcman. MIT Press.

Robbins-Roth, C. 2001. From Alchemy to IPO: The Business of Biotechnology. Perseus


Books,U.S.

Robinson, C. 2008. Exposed: Europe‘s GM-Hype in Times of Food and Fuel Crisis.
Institute of Science in Society http://www.i-sis.org.uk/full/Exposed_GM-
hypeFull.php (Accessed May 8, 2009).

Roe, E.J. 2006. ―Things Becoming Food and the Embodied, Material Practices of an
Organic Food Consumer.‖ Sociologia Ruralis 46:104―121.

Roosevelt, M. 2003. ―Cures On the Cob.‖ Time, May 19


http://www.time.com/time/magazine/article/0,9171,452804-1,00.html (Accessed
May 11, 2009).

Rose, G. 1995. ―Place and Identity: A Sense of Place.‖ in A Place in the World? Places,
Cultures and Globalization, edited by D. Massey and P. Jess.

Said, E. 1975. Beginnings: Intention and Method. Granta.

Said, E. 1995. Orientalism: Western Conceptions of the Orient. New Ed. Penguin Books
Ltd.

Saint-Jore-Dupas, C., L. Faye, and V. Gomord. 2007. ―From planta to pharma with
glycosylation in the toolbox.‖ Trends in Biotechnology 25:317-323.

Sample, I. 2006. ―How a tobacco farm in Kent could provide a life-saving drug for
millions.‖ The Guardian, July 4
http://www.guardian.co.uk/science/2006/jul/04/gm.food.

Schellekens, H. 2004. ―How similar do 'biosimilars' need to be?.‖ Nat Biotech 22:1357-
1359.

329
Schiermeyer, A., S. Dorfmuller, and H. Schinkel. 2004. ―Production of Pharmaceutical
Proteins in Plants and Plant Cell Suspension Cultures.‖ Pp. 91-112 in Molecular
Farming: Plant-made Pharmaceuticals and Technical Proteins, edited by R.
Fischer and S. Schillberg. Wiley VCH.

von Schomberg, R. 2006. ―The precautionary principle and its normative challenges.‖ Pp.
19-42 in Implementing the Precautionary Principle: Perspectives And Prospects,
edited by J. Jones and R. von Schomberg. Edward Elgar Pub.

Seale, C. 2004. Researching Society and Culture. Sage Publications Inc.

Shapin, S. 1988. ―The House of Experiment in Seventeenth-Century England.‖ Isis 79:373-


404.

Shapin, S. 1995. ―Here and Everywhere: Sociology of Scientific Knowledge.‖ Annual


Review of Sociology 21:289―321.

Sismondo, S. 2004. ―Robert K. Merton; Elinor Barber: The Travels and Adventures of
Serendipity: A Study in Sociological Semantics and the Sociology of Science.‖ Isis
95:540.

Smart, A., and P. Martin. 2006. ―The promise of pharmacogenetics: assessing the prospects
for disease and patient stratification.‖ Studies in History and Philosophy of Biol &
Biomed Sci 37:583-601.

Smith, K. E. 2006. ―Problematising power relations in ‗elite‘interviews.‖ Geoforum


37:643-653.

Sonnino, R., and T. Marsden. 2006. ―Beyond the divide: rethinking relationships between
alternative and conventional food networks in Europe.‖ J Econ Geogr 6:181-199.

Sparrow, P., J. Irwin, P. Dale, R. Twyman, and J. Ma. 2007. ―Pharma-Planta: road testing
the developing regulatory guidelines for plant-made pharmaceuticals.‖ Transgenic
Research 16:147―161.

Spök, A. 2007. ―Molecular farming on the rise - GMO regulators still walking a tightrope.‖
Trends in Biotechnology 25:74―82.

Spök, A., R. Twyman, R. Fischer, J. Ma, and P. Sparrow. 2008. ―Evolution of a regulatory
framework for pharmaceuticals derived from genetically modified plants.‖ Trends
in Biotechnology 26:506-517.

Squier, S. M. 2004. Liminal lives: Imagining the human at the frontiers of biomedicine.
Duke University Press.

330
Staeheli, L. 2003. ―Place.‖ Pp. 158-170 in A Companion to Political Geography, edited by
J. Agnew, K. Mitchell, and G. Toal. Wiley-Blackwell.

Star, S. L. 1991. ―Power, technologies and the phenomenology of conventions: on being


allergic to onions.‖ in A Sociology of Monsters: Essays on Power. Technology and
Domination, edited by J. Law. London: Routledge.

Stassart, P., and S.J. Whatmore. 2003. ―Metabolising risk: food scares and the un/re-
making of Belgian beef.‖ Environment and Planning A 35:462, 449.

Stephens, N., P. Atkinson, and P. Glasner. 2008. ―The UK Stem Cell Bank: Securing the
Past, Validating the Present, Protecting the Future.‖ Science as Culture 17:43―56.

Stevenson, F. A., M. Leontowitsch, and C. Duggan. 2008. ―Over-the-counter medicines:


professional expertise and consumer discourses.‖ Sociology of Health & Illness
30:913-928.

Stewart, P., and A. Knight. 2005. ―Trends affecting the next generation of U.S. agricultural
biotechnology: Politics, policy, and plant-made pharmaceuticals.‖ Technological
Forecasting and Social Change 72:521-534.

Stirling, A. 2008. ―"Opening Up" and "Closing Down": Power, Participation, and Pluralism
in the Social Appraisal of Technology.‖ Science Technology Human Values
33:262―294.

Stoger, E., M. Sack, R. Fischer, and P. Christou. 2002. ―Plantibodies: applications,


advantages and bottlenecks.‖ Current Opinion in Biotechnology 13:161-166.

Sturgis, P., and N. Allum. 2004. ―Science in Society: Re-Evaluating the Deficit Model of
Public Attitudes.‖ Public Understanding of Science 13:55-74.

Szerszynski, B. 2005. ―Beating The Unbound: Political Theatre in the Laboratory Without
Walls.‖ in Performing Nature: Explorations in Ecology and the Arts, edited by G.
Giannachi and N. Stewart. Frankfurt and New York: Peter Lang.

Tacket, C. et al. 1998. ―Immunogenicity in humans of a recombinant bacterial antigen


delivered in a transgenic potato.‖ Nat Med 4:607-609.

Thrift, N. 2006. ―Re-inventing invention: new tendencies in capitalist commodification.‖


Economy and Society 35:279-306.

Tutton, R. 2007. ―Constructing Participation in Genetic Databases: Citizenship,


Governance, and Ambivalence.‖ Science Technology Human Values 32:172-195.

331
Twyman, R. M., S. Schillberg, and R. Fischer. 2005. ―Transgenic plants in the
biopharmaceutical market.‖ Expert Opinion on Emerging Drugs 10:185-218.

Vallely, P. 2009. ―Strange fruit: Could genetically modified foods offer a solution to the
world's food crisis?.‖ The Independent, April 18 http://www.independent.co.uk/life-
style/food-and-drink/features/strange-fruit-could-genetically-modified-foods-offer-
a-solution-to-the-worlds-food-crisis-1668543.html (Accessed April 23, 2009).

Van Lente, H. 2000. ―From promises to requirement.‖ in Contested Futures: A Sociology of


Prospective Techno-science, edited by N. Brown, B. Rappert, and A. Webster.
Ashgate.

Van Reenen, J. 2002. ―Economic issues for the UK biotechnology sector.‖ New Genetics
and Society 21:109-130.

Vidler, E., and J. Clarke. 2005. ―Creating Citizen-Consumers: New Labour and the
Remaking of Public Services.‖ Public Policy and Administration 20:19-37.

Wainwright, S., and C. Williams. 2008. ―Spaces of speech and places of performance: an
outline of a geography of science approach to embryonic stem cell research and
diabetes.‖ New Genetics and Society 27:161―173.

Wainwright, S., C. Williams, M. Michael, B. Farsides, and A. Cribb. 2006. ―From bench to
bedside? Biomedical scientists' expectations of stem cell science as a future therapy
for diabetes.‖ Social Science & Medicine 63:2052―2064.

Waldby, C. 2000. The visible human project: Informatic bodies and posthuman medicine.
Routledge.

Walsh, G. 2007. Pharmaceutical biotechnology: Concepts and Applications. London:


Wiley-Blackwell.

Walsh, Gary. 2003. Biopharmaceuticals. John Wiley and Sons.

Waterton, C., and B. Wynne. 1999. ―Can focus groups access community views?.‖ Pp. 127-
143 in Developing Focus Group Research: Politics, Theory and Practice, edited by
J. Kitzinger and R. Barbour. Sage Publications Ltd.

Webster, A., C. Douglas, and G. Lewis. 2009. ―Making Sense of Medicines: ‗Lay
Pharmacology‘ and Narratives of Safety and Efficacy.‖ Science as Culture 18:233-
247.

Whatmore, S. 2002. Hybrid Geographies: Natures Cultures Spaces: Natures, Cultures,


Spaces. London: Sage Publications Ltd.

332
WHO. 2007. Rabies Vaccine Position Paper.
http://www.who.int/immunization/documents/positionpapers/en/index.html
(Accessed May 10, 2009).

Williams, S., J. Gabe, and P. Davis. 2008. ―The sociology of pharmaceuticals: progress and
prospects.‖ Sociology of Health & Illness 30:813-824.

Wilsdon, J., and R. Willis. 2004. ―See-through Science: Why public engagement needs to
move upstream.‖.

Withers, C.W.J. 2002. ―Nature's Government: Science, Imperial Britain, and the
"Improvement" of the World.‖ Journal of Historical Geography 28:285-286.

Wright, S. 1994. Molecular Politics: Developing American and British Regulatory Policy
for Genetic Engineering, 1972-82. University of Chicago Press.

Wyatt, S., and B. Balmer. 2007. ―Home on the Range: What and Where is the Middle in
Science and Technology Studies?.‖ Science Technology Human Values 32:619-626.

Wynne, B. 1991. ―Knowledges in Context.‖ Science, Technology, and Human Values


16:121, 111.

Wynne, B. 1992. ―Misunderstood misunderstanding: social identities and public uptake of


science.‖ Public Understanding of Science 1:304, 281.

Wynne, B. 1996. ―May the sheep safely graze? A reflexive view of the expert-lay
knowledge divide.‖ Risk, Environment and Modernity: Towards a New Ecology 44–
83.

Wynne, B. 2001. ―Creating Public Alienation: Expert Cultures of Risk and Ethics on
GMOs.‖ Science as Culture 10:481, 445.

Wynne, B. 2002. ―Risk and Environment as Legitimatory Discourses of Technology:


Reflexivity Inside Out?.‖ Current Sociology 50:459―477.

333

Вам также может понравиться