the glivec precedent

Upload: soham-mukherjee

Post on 23-Feb-2018

226 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/24/2019 The Glivec Precedent

    1/20

    Special Issue on The GlivecPrecedent, Economic and

    Political Weekly, Vol.

    XLVIII No.32, pages 4157.

  • 7/24/2019 The Glivec Precedent

    2/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 41

    Who Invented Glivec?Does It Matter Anyway?

    Graham Dutfield

    This article looks at Glivecs

    journey from its invention to

    its patenting and sale while

    questioning the concept of credit

    for inventions in science and

    technology.

    In 2003, the then Novartis Chief

    Executive Officer Daniel Vasella pub-

    lished a book (Vasella and Slater

    2003) in praise of Glivec detailing his

    firms major role in delivering this un-

    commonly effective life-saving medi-

    cine. But, who was really responsible? In

    the original USpatent,1one single per-

    son is named as the inventor: Jrg Zim-

    mermann of Ciba Geigy (later Novartis).

    In the later USpatent on the beta crystal-

    line form, held in India to be unpatentable,

    Zimmermann is joined by two Novartis

    colleagues.2However, contrary to what

    this might suggest, chronic myeloid

    leukaemia (CML) patients would never

    have received a product called Glivec if

    they had had to rely solely on Novartis.

    Credit in science and technology is a

    devilishly tricky matter to be objective

    about. Effort, dedication, inspiration, vision,

    ambition, intuition, counter-intuition, ad-

    vanced technical knowledge in one or moredisciplines, skill, serendipity, networking

    abilities, and sheer bloody-mindedness,

    all help make a creative achievement

    possible. But, assessing what contributions

    and which people are not just important,

    but indispensable, can be a tough task.

    The various Glivec stories bring to-

    gether a whole cast of actors and locations

    over broad geographical and chronologi-

    cal distances (Brody 2007: 13-18; Keating

    and Cambrosio 2012; Mukherjee 2010:

    430-43; Nathan 2007: 180-85). Medicines

    have histories and Glivec is no exception,

    being the final destination of a long and

    winding historical learning trail; one

    that begins in 1960 with a foundational

    discovery by two US-based scientists of a

    chromosomal defect, which they suggested

    had a causal relationship with chronic

    granulocytic leukemia (Nowell and

    Hungerford 1960: 1497).

    Glivec: To Whose Credit?

    Which stories are most complete and

    accurate depends largely on what Glivec

    is. Glivec, the synthetic small molecule,

    a pyrimidine derivative generically known

    as imatinib, is one thing. If Zimmermann

    was the first and only person to make

    this molecule, he is rightly the inventor of

    it as long as we are unconcerned about

    scientific credit and merely need to

    identify one or a few individuals closelyassociated with its first manufacture.

    But, Glivec is much more than this or

    that pyrimidine derivative in this or that

    structural form. It is a pharmaceutical

    product of a type known as a tyrosine

    kinase inhibitor; one that is extraordi-

    narily effective in the treatment of CML,

    and that was made to a specification

    well worked out by others. Potentially,

    an almost infinite variety of different

    molecules can be made in the laboratory.

    But, to identify a class of chemicals to be

    made for a specific purpose, make them,

    analyse them, select a few for testing, test

    them in various ways, select the best one,

    optimise it, and finally get it approved for

    sale, requires the contribution of many

    more people than a synthetic chemist

    working in a lab, however talented.

    One way to look fairly at credit is to

    ask which people were the sine qua non,

    those without whom there would have

    been no Glivec. The importance of theindividual coming up with the actual

    chemical cannot be underestimated, but

    the inventor named on the patent can

    also be seen in such cases as the one who

    added the finishing touches, inching the

    idea forward over the novelty, inventive

    step and industrial applicability thresh-

    olds. Legally, it is immaterial whether

    this persons effort or intellectual input

    was proportionately significant compared

    to anybody elses, and this may not

    necessarily be seen as unfair.

    None of the numerous scientists who

    contributed has publicly challenged

    Zimmermanns sole inventorship. Zimmer-

    mann did spend more than two years in

    developing the right molecule and then

    preparing it in a form that could be taken

    orally. Designing the last piece of the

    jigsaw was no quick and easy task (Buch-

    dunger and Zimmermann ND).

    Competing claims could be made for

    Nicholas Lydon or Alex Matter of CibaGeigys programme on kinase inhibitors.

    But, if one really can single out the

    Graham Dutfield ([email protected])

    teaches International Governance at the

    School of Law, University of Leeds, UK.

  • 7/24/2019 The Glivec Precedent

    3/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly42

    irreplaceable person without whom

    there would have been neither an inven-

    tion nor a pharmaceutical product called

    Glivec, then it would probably be Brian

    Druker, initially of the Dana-Farber Can-

    cer Institute, and thereafter at the Ore-

    gon Health Sciences University.

    From Brian Druker to Novartis

    Druker was not just lead author of the

    article that announced Glivec to the

    world under the name of CPG57148

    (Druker et al 1996). He was relentless in

    his determination to find a treatment

    for CML and to make it available to as

    many patients as possible. This com-

    mitment to do all that was necessary to

    cure leukaemia patients led him to

    collaborate with Lydon and Ciba Geigy

    to harness the companys biochemical

    capacity in kinase inhibition to his

    search for a molecule that was able

    specifically to block the action of a sin-

    gle aberrant enzyme common to the

    cells of CMLsufferers. His role did not

    end when the molecule was discovered

    and, largely under his direction, shown

    to work.

    In an interview with author-medical

    scientist Siddhartha Mukherjee (2010:

    436), Druker described his incrediblypersistent efforts to talk a reluctant

    Novartis, concerned about the small

    number of patients and, therefore, the

    lack of a market for the drug, into

    producing enough for clinical trials.

    Ironically, the specificity of Glivec that

    made it such an outstanding drug did

    not favour it as a commercial product

    from Novartiss perspective: it kept the

    patient base small.

    In the event, Novartis went ahead

    with Glivec, which turned out to be to

    its considerable advantage. In May 2001,

    in unusually quick time, the USFood and

    Drug Administration approved Glivec.

    Novartis subsequently set a global

    patient-per-month price of $2,400. Even

    with patient assistance for poorer peo-

    ple, which enabled some to get it for

    free, the drug proved to be highly profit-

    able. Besides, such programmes, bene-

    ficial as they obviously are to some

    patients, are an excellent way to dis-courage generic market entry where

    there are no patents. By 2003, annual

    sales had reached $1 billion, and reve-

    nues have risen considerably since then.

    True to his convictions, Druker was

    one of over 100 physicians who recently

    publicly criticised the high prices of

    drugs for CML(Experts in Chronic Mye-

    loid Leukemia 2013). Such an article

    could have been published several yearsago: this is hardly a new problem. But,

    one hopes at this late stage that while

    the key patents are still in force in sev-

    eral countries, and the original USpat-

    ent having been extended from its expi-

    ry date of 28 May 2013 for 586 addition-

    al days, it might make a difference. It is

    also worth noting that Druker report-

    edly welcomed the Indian Supreme

    Court decision.

    Brian Drukers name may not be on

    any of the patents. But, it would be

    hard to name any other individual to

    whom leukaemia patients should be

    more grateful.

    Notes

    1 US Patent no 5521184, Pyrimidine Derivativesand Processes for the Preparation Thereof,28 May 1996.

    2 US States Patent no 6894051, Crystal Modi-fication of a N-phenyl-2-Pyrimidineamine

    Derivative, Processes for Its Manufacture andIts Use, 17 May 2005.

    References

    Brody, H (2007):Hooked: Ethics, the Medical Profes-sion and the Pharmaceutical Industr y (Lanham:Rowman & Littlefield).

    Buchdunger, E and J Zimmermann (nd): The

    Story of Gleevec, viewed on 2 June 2013,http://www.innovation.org/index.cfm/StoriesofInnovation/InnovatorStories/The_Story_of_Gleevec

    Druker, B, S Tamura, E Buchdunger, S Ohno,G M Segal, S Fanning, J Zimmermann andN B Lydon (1996): Effects of a Selective Inhibitorof the Abl Tyrosine Kinase on the Growth ofBcr-Abl Positive Cells, Nature Medicine , 2(5):561-66.

    Experts in Chronic Myeloid Leukemia (2013): ThePrice of Drugs for Chronic Myeloid Leukemia(CML) is a Reflection of the UnsustainablePrices of Cancer Drugs: From the Perspective ofa Large Group of CML Experts,Blood, 121(22):4439-42.

    Keating, P and A Cambrosio (2012):Cancer on Trial:Oncology as a New Style of Practice

    (Chicago:University of Chicago Press).

    Mukherjee, S (2010): The Emperor of All Maladies:A Biography of Cancer (New York: Scribner).

    Nathan, D G (2007): The Cancer Treatment Revolu-tion: How Smart Drugs and Other NewTherapies Are Renewing Our Hope and Chang-ing the Face of Medicine (Hoboken: John Wiley& Sons).

    Nowell, P C and D A Hungerford (1960): A MinuteChromosome in Human Chronic GranulocyticLeukemia, Science, 132(3438): 1497.

    Vasella, D and R Slater (2003):Magic Cancer Bullet:How a Tiny Orange Pill Is Rewrit ing MedicalHistory (New York: HarperBusiness).

    REVIEW OF WOMENS STUDIES

    May 4, 2013

    Intersections of Gender and Caste Sharmila Rege, J Devika, Kalpana Kannabiran,

    Mary E John, Padmini Swaminathan, Samita Sen

    Revitalising Dalit Feminism: Towards

    Reflexive, Anti-Caste Agency of Mang

    and Mahar Women in Maharashtra Smita M Patil

    Caste and Gender in a Mumbai Resettlement Site Varsha AyyarDalit Women as Political Agents: A Kerala Experience Rekha Raj

    The Mathammas: Gender, Caste and the Politics

    of Intersectionality in Rural Tamil Nadu Anandhi S

    The Concept of Honour: Caste Ideology

    and Patriarchy in Rural Maharashtra Manisha Gupte

    Cultural Gandhism: Casting Out the Dalit Woman Swathy Margaret

    Ruptures and Reproduction in Caste/Gender/Labour Meena Gopal

    For copies write to: Circulation Manager,

    Economic and Political Weekly,

    320-321, A to Z Industrial Estate, Ganpatrao Kadam Marg,

    Lower Parel, Mumbai 400 013.email: [email protected]

  • 7/24/2019 The Glivec Precedent

    4/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 43

    Two Decades of Struggle

    Amit Sengupta

    The Supreme Court judgment in

    the Novartis case is important as

    it vindicates the entire process

    leading to health safeguards

    being incorporated in the Indian

    Patents Act. The article discusses

    this process, from the General

    Agreement on Tariffs and Trade

    and popular mobilisation in

    India to the enactment of and

    amendments to the Act, in thebackdrop of the judgment.

    The judgment by the Supreme Court

    of India, denying the claim of apatent on the anti-leukaemia drug

    Glivec (imatinib) by the Swiss multi-

    national Novartis, is important at many

    levels. In this article we discuss, in the

    backdrop of the judgment, the long and

    protracted course leading to the enact-

    ment of the Indian Patents Act of 2005.

    The Uruguay Round

    In 1986, a new round of negotiations was

    initiated under the General Agreement

    on Tariffs and Trade (GATT), otherwise

    known as the Uruguay Round of negoti-

    ations. In the Uruguay Round, developed

    countries introduced a number of issues

    on the agenda which were hitherto

    not considered trade issues related to

    intellectual property (IP) rights, invest-

    ment and services.

    Initially, developing countries led by

    India and Brazil were able to stall the

    introduction of these new issues (Shukla

    2000: 14-15), while the UScontinued topress for their inclusion. The latters

    position was dictated by the state of the US

    economy. Having lost its competitive edge

    in the manufacturing sector and with its

    own agricultural exports threatened by

    state-subsidised agricultural exports from

    Europe, the USwas keen to open up the

    services sector especially for financial

    services. At the same time, the UShad an

    interest in protecting its IP-dependent

    industries where it still had an advantage,

    specifically in pharmaceuticals, software

    and audiovisual media (ibid: 20-21).

    India had a clear interest in not

    agreeing to these new demands. Indias

    pharmaceutical sector had flourished in

    the wake of its 1970 Patents Act, which did

    not allow product patents on medicines

    and agro-chemicals. India only allowed

    process patents on pharmaceuticals, and

    had leveraged on this to develop capacity

    in process technologies.

    By the beginning of 1989, the resistanceby developing countries was broken down.

    Enormous pressure exerted by the US

    resulted in the two main hold-outs

    changing their position. India went to

    the extent of replacing Indias chief

    negotiator at GATT, S P Shukla, because

    of his strong opposition to the inclusion

    of IP issues in the negotiating agenda

    (Marcellin 2010: 87).

    The significance of the negotiations wasnot clear to most popular movements and

    civil society groups in different parts of

    the world. A key to the development of

    the resistance in India was the formation

    of the National Working Group on Patent

    Laws (NWGPL). In spite of its relatively

    small numbers, the NWGPLwas hugely

    influential in shaping opposition to the

    Trade-Related Aspects of Intellectual

    Property Rights (TRIPS) Agreement, right

    from the late 1980s. It was composed of

    a group of former civil servants, lawyers,

    scientists, representatives of the domestic

    pharmaceutical industry and represent-

    atives of trade unions in the pharma-

    ceutical industry.1

    The NWGPL, itself not a mass movement,

    became a catalyst for advocacy and

    mobilisation. It was the principal source of

    evidence-based arguments against the

    proposed regime on IP. Strong support

    from the domestic industry found reso-

    nance among a wide range of politicalactors. Over the next decade, the NWGPL

    organised the Forum of Parliamentarians,

    which had representation from virtually the

    entire political spectrum. Several political

    and social movements, non-governmental

    organisations and mass organisations in

    India formed alliances against the GATT

    negotiations. Many subsequent develop-

    ments had their roots in the popular mo-

    bilisations between 1990 and 2005.

    Tortuous Path

    The path towards the final formulation

    of Indias Patents Act was also increas-

    ingly informed by, from 1991, the formal

    introduction of neo-liberal reforms. From

    an earlier position that India was forced

    to concede to in the GATTnegotiations,

    there was now an attempt to argue that

    strong IP protection would promote

    domestic interests. However, popular

    sentiment continued to be hostile.

    The TRIPSAgreement provided a three-stage time framework for developing

    countries: introduction of a mailbox

    Amit Sengupta ([email protected])

    is co-convenor of Jan Swasthya Abhiyan

    (Peoples Health Movement, India) and is alsoassociated with the All India Peoples Science

    Network.

  • 7/24/2019 The Glivec Precedent

    5/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly44

    facility and Exclusive Marketing Rights

    (EMRs) from 1995; provisions on rights

    related to term of patent protection,

    compulsory licensing, reversal of burden

    of proof, etc, by 2000; and introduction

    of product patent protection in all fields

    from 1 January 2005.

    The political instability in India, post-1996, meant that further discussions on

    amendments to Indias 1970 Act resumed

    only in 1998 after the installation of the

    Bharatiya Janata Party (BJP)-led National

    Democratic Alliance (NDA) government.

    Indian Parliament enacted two legislations

    through the Patents (Amendment) Act of

    1999 and 2002, which addressed the first

    two requirements of the TRIPSAgreement.

    After assuming office, the NDAgovern-

    ment was clearly subsumed by the neo-

    liberal logic while engaging with public

    policy on a range of issues.2 The NDA

    government then circulated the draft

    Third Patents (Amendment) Bill in 2003,

    but it could not be discussed because of

    the change of government in 2004.

    In 2004, there was a clear consensus

    between the two principal parties in India

    the Congress and the BJP and the

    United Progressive Alliance (UPA) govern-

    ment circulated an almost unchanged

    version of the NDAs Third Patents

    (Amendment) Bill draft. In the then

    political spectrum only the left parties

    (along with some regional parties) stood

    firmly against the draft Bill. But towards

    the end of 2004, the BJPstarted voicing

    opposition to the draft Bill. While this is in

    the realm of speculation, BJPs volte-face

    had little to do with any opposition to thesubstance of the Bill (given that this was

    identical to the Bill they had circulated)

    and more to do with an intent to embarrass

    the UPA government. With support for

    the bill now unsure, the UPAgovernment

    decided to beat the 31 December 2004

    deadline by promulgating an ordinance

    on 26 December 2004 (The Patents

    (Amendment) Ordinance 2004).

    Patents Ordinance of 2004

    The Ordinance, if ratified by Parliament,

    would have made it impossible for Indian

    companies to continue producing cheaper

    versions of new drugs. In early 2005, with

    the BJPengaged in a bitter tussle with the

    Congress in Bihar and Jharkhand over

    formation of ministries, it became clear

    that the Ordinance would be defeated in

    Parliament and the Congress was now

    forced to seek the lefts support.

    In the consequent negotiations between

    the left and the government, the left

    largely depended on advice provided by

    people associated with the NWGPL. These

    negotiations also allowed other interest-

    ed parties to suggest new language. At

    the end, several important amendments

    were made to the 2004 Ordinance

    (ICTSD2005), including the insertion of

    Section 3(d), which has been the subjectof much discussion after its use by the

    Supreme Court to strike down the

    appeal by Novartis.

    The negotiations were held in the back-

    drop of protests across the country, as

    well as in different parts of the world

    all demanding that the pharmacy of the

    South should not be jeopardised. By 2005,

    the global Access to Medicines campaign

    was a powerful force and organisations

    such as Mdecins Sans Frontires (MSF)

    and others were able to organise support

    across the globe. Protest letters were

    sent to the prime minister, including one

    where the co-signatories included Jim

    Yong Kim, the present World Bank chief

    (then director, Department of HIV/AIDS,

    World Health Organization) (Khor 2013).

    Important Amendments

    While there has been considerable

    focus on Section 3(d) of the amended

    Act, many important amendments

    NEW

    Pp xiv + 538 Rs 745

    ISBN 978-81-250-5131-2

    2013

    Higher Education in IndiaIn Search of Equality, Quality and Quantity

    Edited by

    JANDHYALAB G TILAK

    India has a large network of universities and colleges with a massive geographical reach and the facilities for higher

    education have been expanding rapidly in recent years. The story of higher education in India has seen many challenges

    over the decades and has not been without its share of problems, the most serious being a very high degree of inequity.

    Drawn from writings spanning almost four decades in the EPW, the articles in this volume discuss, among other things,

    issues of inclusiveness, the impact of reservation, problems of mediocrity, shortage of funds, dwindling numbers of faculty,

    and unemployment of the educated young.

    Authors: Andr Bteille Shiv Visvanathan Suma Chitnis Satish Deshpande K Sundaram Rakesh Basant, Gitanjali Sen Jayati Ghosh

    Thomas E Weisskopf Lloyd I Rudolph, Susanne Hoeber Rudolph A M Shah Errol DSouza G D Sharma, M D Apte Glynn L Wood

    D P Chaudhri, Potluri Rao R Gopinathan Nair, D Ajit D T Lakdawala, K R Shah Chitra Sivakumar Amrik Singh Jandhyala B G Tilak

    Anindita Chakrabarti, Rama Joglekar Karuna Chanana Saumen Chattopadhyay Samuel Paul Deepak Nayyar V M Dandekar M Anandakrishnan

    Thomas Joseph

    Orient Blackswan Pvt Ltdwww.orientblackswan.com

    MumbaiChennai New Delhi KolkataBangaloreBhubaneshwar Ernakulam GuwahatiJaipurLucknow Patna Chandigarh HyderabadContact: [email protected]

  • 7/24/2019 The Glivec Precedent

    6/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 45

    to the 2004 Ordinance were adopted,

    including:

    (1) Restrictions on Patentability: The

    amendments clarified that an inventive

    step means a feature of an invention that

    involves technical advances as compared

    to the existing knowledge or having eco-nomic significance or both. It incorpo-

    rated a new definition for new invention:

    any invention or technology which has not been

    anticipated by publication in any document

    or used in the country or elsewhere in the world

    before the date of filing of patent application

    with complete specification, i e, the subject

    matter has not fallen in public domain or that

    it does not form part of the state of the art.

    It also provided a definition forpharma-

    ceutical substanceas beinga new entity

    involving one or more inventive steps.

    (2) Restoration of Pre-grant Opposition

    to Patents: The amendments restored

    all the original grounds in the previous

    Act for opposing grant of a patent and

    also provided that: the Controller shall,

    if requested by such person for being

    heard, hear him. The time for filing

    such opposition was extended from

    three to six months.

    (3) Export to Countries Without Manu-facturing Ability: The amendments

    clarified that a country could import

    from India if it by notification or other-

    wise allowed importation of the patent-

    ed pharmaceutical product from India.

    (4) Continued Manufacture of Drugs

    with Applications in Mailbox: The

    amendments clarified that Indian com-

    panies that were already producing drugs

    that were the subjects of mailbox applica-

    tions could continue to produce them

    after payment of a royalty, even if the

    drug was subsequently granted a patent.

    (5) Time Period for Considering Com-

    pulsory Licence Application: Concerns

    that the process of granting compulsory

    licences could take too long were ad-

    dressed by specifying that the reasonable

    time period before the Patents Controller

    considers issuance of a compulsory licence

    when such a licence is denied by thepatent holder shall not ordinarily exceed

    six months.

    (6) Export by Indian Companies of

    Patented Drugs: The amendments pro-

    vided that when patented drugs are

    produced under compulsory licence in

    India the licensee may also export the

    patented product.

    Several of the amendments are being

    used today by different groups to try tosafeguard access. In particular, the pre-

    grant opposition provisions have been

    used extensively by domestic companies

    and civil society groups, and combined

    with restrictions on patentability, the

    provisions have allowed many important

    drugs to be kept off patents. Further,

    a number of drugs introduced in the

    transition phase (1995-2005) were not

    patented as the amended Act allowed

    generic companies to manufacture and

    sell drugs introduced in this period.

    The language for Section 3(d) was

    provided by the Indian Drug Manufac-

    turers Association (IDMA). The left parties

    had asked for a more stringent definition

    of patentability by limiting grant of patents

    for pharmaceutical substances to new

    chemical entities or to new medical

    entities involving one or more inventive

    steps. Section 3(d) was a compromise

    and the government had agreed to refer

    the matter to an expert panel.Subsequently, the government consti-

    tuted a Technical Expert Group under

    the chairmanship of R A Mashelkar,

    former director general, Council of

    Scientific and Industrial Research. The

    Group, in its report in 2007, opined that

    restriction of patents to new chemical

    entities would be incompatible with the

    TRIPSAgreement. Evidence surfaced that

    parts of the report had been plagiarised

    from a study by the UK-based Intellectual

    Property Institute, funded by Interpat,

    an association of 29 drug companies

    including Novartis (Padma 2007: 392).

    The report was withdrawn and press

    reports indicated that Mashelkar had

    resigned from the committee (ibid). Yet,

    the same committee resubmitted a new

    version with the same conclusions in

    2009. These recommendations were ex-

    peditiously accepted by the government.

    Vindication of Struggle

    The Supreme Court judgment in the

    Novartis case, thus, needs to be read not

    just as an instance of the application of

    one section (Section 3(d)) of the Indian

    Patents Act. The judgment is important

    as it vindicates the entire process that

    led to health safeguards being incorpo-

    rated in the Indian Act.

    The judgment, in fact, refers clearly to

    this process by noting (in para 26):3

    to understand the import of the

    amend-

    ments in clauses (j) and (ja) of section 2(1)

    and the amendments in section 3 it is

    neces-

    sary to find out the concerns of Parliament,

    based on the history of the patent law in

    the

    country, when it made such basic changes in

    the Patents Act. What were the issues the

    legislature was trying to address? What was

    the mischief Parliament wanted to check

    and

    what were the objects it intended to

    achieve through these amendments?

    The judgment is a vindication not

    just of a legislative process, but of popu-

    lar resistance and mobilisation in

    India and across the world that chal-

    lenged corporate power. Small victories

    such as this become inspirations for

    larger battles.

    Notes

    1 For more informat ion about the formation ofthe NWGPL, see Sen Gupta (2010).

    2 See, for example, Arulanantham (2004).

    3 Text of final judgment is available at: http://ju-

    dis.nic.in/supremecourt/imgs1.aspx? filename=40212 (viewed on 20 June 2013).

    References

    Arulanantham, David P (2004): The Paradox ofthe BJPs Stance Towards External EconomicLiberalisation: Why a Hindu Nationalist PartyFurthered Globalisation in India, Asia Pro-gramme Working Paper, December, RoyalInstitute of International Affairs, ChathamHouse, London.

    ICTSD (2005): Indian Parliament Approves Con-troversial Patent Bill, Bridges Weekly Trade

    News Digest, 9(10), International Centre forTrade and Sustainable Development, viewedon 20 June 2013, http://ictsd.org/i/news/

    bridges weekly/7294/Khor, Martin (2013): A Victory for Patients Access

    to Medicines, Global Trends Series, ThirdWorld Network, 8 April, viewed on 20 June2013, http://www.twnside.org.sg/title2/gtrends/2013/gtrends426.htm

    Marcel lin, Sherr y S (2010):The Political Economy ofPharmaceutical Patents: US Sectional Interestsand the African Group at the WTO(Farnham,England: Ashgate Publishing).

    Padma, T V (2007): Plagiarised Report on PatentLaws Shames Indian Scientists,Nature Medicine,13(4): 392.

    Sen Gupta, Amit (2010): B K Keayla: A PersonalReminiscence, Economic & Political Weekly,45(51): 25-26.

    Shukla, S P (2000): From GATT to WTO and Be-yond, Working Papers No 195, United NationsUniversity/World Institute for DevelopmentEconomics Research, Helsinki, Finland.

  • 7/24/2019 The Glivec Precedent

    7/20

  • 7/24/2019 The Glivec Precedent

    8/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 47

    Chennai Patent Controller, and was kept

    in the mailbox.

    Hit by Section 3(d)

    After 1 January 2005, the mailbox facility

    was opened. Five pre-grant oppositions

    were filed, one by the Cancer Patients Aid

    Association (CPAA), and four by Indiangeneric companies.9Novartis contended

    that the claimed invention was novel,

    inventive and industrially applicable. The

    oppositionists contended otherwise. They

    also argued that the claimed invention

    was hit by Section 3(d) of the Patents Act.

    In this respect, Novartis had filed ad-

    ditional affidavits ostensibly to show

    that on account of the alleged increased

    bioavailability (30%) of the crystalline

    form imatinib mesylate, there was a sig-

    nificant increase in the efficacy in the

    claimed invention.10While there was no

    dispute as to the claimed inventions

    industrial applicability, the Patent Con-

    troller concluded that the claimed in-

    vention was neither novel nor inventive

    and was also hit by Section 3(d), thus,

    rejecting the application.

    Against this order, Novartis filed ap-

    peals before the Madras High Court.

    These were later transferred to the Intel-

    lectual Property Appellate Board (IPAB).Both, NovartisAGand Novartis India also

    challenged the validity of Section 3(d) on

    the grounds that it did not comply with

    the TRIPSAgreement and the term effi-

    cacy was vague and, therefore, in viola-

    tion of Article 14 of the Constitution. The

    Madras High Court dismissed both the

    Writ Petitions on both the grounds. It also

    held that Section 3(d) had been enacted,

    amongst others reasons, to protect public

    health, observing,

    We have borne in mind the object which the

    amending Act wanted to achieve namely, to

    prevent evergreening; to provide easy access

    to the citizens of the country to life saving

    drugs and to discharge their constitutional

    obligation of providing good health care to

    its citizens.11

    Importantly, Novartis did not challenge

    the order of the Madras High Court.

    In the appeals, the IPABreversed the

    Patent Controller order on two grounds,

    and held that the claimed invention was

    novel and inventive. However, it agreedwith the Patent Controller that it was hit

    by Section 3(d) and rejected the patent

    application. It was against this order

    that Novartis filed the Special Leave

    Petition in the Supreme Court.

    In the Supreme Court, the maintain-

    ability of Novartiss appeal was dropped

    and it was heard on merits.

    On MeritsNovartis had patented imatinib, which

    was exemplified in Example 21 of the

    Zimmerman patent. Novartis argued

    that the claimed invention involved a

    twofold step over Zimmerman, firstly

    from imatinib to imatinib mesylate, the

    salt form, and secondly to the crystal-

    line form of imatinib mesylate.

    On the interpretation of Section 2(1)(j)

    and (ja) and Section 3, the Court, speak-

    ing through Justice Aftab Alam, held that

    if they are read together, even though a

    product or a process may satisfy the test

    of invention under 2(1)(j) and (ja), it

    may still be held not patentable under

    Section 3 of the Act.

    On Novelty

    The Court noted that the salt form was

    actually claimed in the Zimmerman ap-

    plication itself. Moreover, the acid addi-

    tion salt forms could be made in a cus-

    tomary manner or in the manner knownper se. That apart, Novartis itself had

    made a Patent Term Extension Application

    in the US in the Zimmerman patent, in

    respect of the mesylate form. This was

    granted for a period of 586 days on the

    basis that the Zimmerman patent covered

    the mesylate form. Also, Novartis had

    sought an injunction against Natco in

    the UKfrom marketing the mesylate form.

    Therefore, the Court observed, that

    there was no room for doubt that imat-

    inib mesylate, marketed as Glivec, was

    submitted for drug approval and was

    covered by the Zimmerman patent.

    The Court also observed that the

    properties of imatinib, namely the in-

    hibition of tyrosine kinase activity, were

    also found in the pharmaceutically

    acceptable salt forms, particularly not-

    ing that the authors of Cancer Research

    had concluded that in respect of proper-

    ties no significant difference in results

    could be seen between the two forms ofCGP 57148.12 Similar findings were

    recorded in an article in the journal,

    Nature. The Court, therefore, concluded

    that it was unable to see how Imatinib

    Mesylate can be said to be a new product,

    and was in fact covered by the

    Zimmerman patent.

    In order to get over this hurdle,

    Novartis admitted that though imatinib

    mesylate may have been claimed andtherefore covered under the Zimmerman

    patent, it was not disclosed, as there was

    no enabling disclosure in the Zimmerman

    patent. Novartis relied on the decision of

    the US court of Customs and Patent

    Appeals inHogan.13The Court found that

    Hogan was rendered in very specific

    circumstances and later decisions of the

    US courts had narrowed that down to

    Hogans impact. Additionally, on the basis

    that the artificial distinction between

    coverage and disclosure negated the fun-

    damental rule underlying patents, the

    argument was rejected.

    The Crystalline Form:

    Interpreting 3(d)

    For the purposes of argument the Court

    accepted that the crystalline form im-

    atinib mesylate was new and not obvi-

    ous.14Of course, the crucial question be-

    fore the Court was, whether in view of

    Section 3(d) as amended, Novartis couldbe granted a patent on the claimed in-

    vention of the crystalline form of imat-

    inib mesylate.

    The Court observed that in order to

    correctly understand the present law it

    would be necessary to briefly delve into the

    legislative history of the law of patents in

    the country. It noted the parliamentary

    debate during the final amendments to

    the Patents Act in 2005, with concerns

    about evergreening in pharmaceutical

    patents. It recalled that Section 3(d) is

    directed at these practices.

    In interpreting the new provisions,

    the Court held that the Act provided for

    the duality of the concepts of invention

    and patentability. Not all inventions

    under the Act were patentable, as is

    illustrated by Sections 3 and 4. The Court

    rejected the submission by Novartis that

    Section 3(d) was trifling change. It held

    that different standards are set by the

    Act for medicines and other chemicalsubstances and that Section 3(d) had

    been enacted to prevent evergreening.

  • 7/24/2019 The Glivec Precedent

    9/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly48

    The Court found that as the crystal-

    line form imatinib mesylate was a new

    form of imatinib mesylate, Section 3(d)

    would apply.

    However, Novartis argued that neither

    imatinib nor imatinib mesylate had

    known efficacy. Therefore, no compari-

    son could be made of the crystallineform of imatinib mesylate with a known

    substance with known efficacy as

    required under Section 3(d). This was re-

    jected by the Court on the grounds that it

    is well established by the Supreme Court

    itself, as interpreted in Monsanto,15 that

    the expression publicly known was

    held not to mean that it was widely used

    to the knowledge of the consumer pub-

    lic, but that it is sufficient if it is known

    to the persons who are engaged in the

    pursuit of knowledge of the patented

    product or process either as men of sci-

    ence or men of commerce or consumers.

    In examining the efficacy of the dif-

    ferent forms of imatinib, that is imatinib

    free base, non-crystalline form of imat-

    inib mesylate, and the crystalline form

    of imatinib mesylate, the Court noted that

    it was Novartiss own case that all the

    properties possessed by the imatinib free

    base were possessed by the crystalline

    form of imatinib mesylate. In the circum-stances the Court queried how there

    could be any enhanced efficacy in the

    crystalline form of imatinib mesylate?

    In this respect, Novartis filed two affi-

    davits to satisfy the requirements of

    Section 3(d) for consideration.

    One of these affidavits stated that on

    conducting experiments it was found

    that there was a 30% increase in bio-

    availability in the crystalline form of

    imatinib mesylate as compared to the

    imatinib free base. The Court noted that it

    was Novartiss own case that the product

    immediately preceding the crystalline

    form imatinib mesylate is the non-

    crystalline form of imatinib mesylate.

    The non-crystalline form of imatinib

    mesylate was thus known. The Court

    found that the comparison with imatinib

    free base was inappropriate.

    The Court therefore held that Novartis

    was bound to show enhanced efficacy

    of crystalline imatinib mesylate overnon-crystalline imatinib mesylate, which

    Novartis had failed to do. Moreover, the

    Court opined that the bioavailability

    of the crystalline imatinib mesylate

    would be on account of the salt form,

    that is the non-crystalline form of

    imatinib mesylate.

    Efficacy Is Therapeutic Efficacy

    The Court held that the term efficacy in3(d) has to be understood as the ability

    to produce the desired or intended

    effect. This would differ in the context

    of the product, its function, utility or the

    purpose under consideration. In the con-

    text of medicines that claim to cure a

    disease, it has to be therapeutic efficacy.

    Considering the context of 3(d), the

    Court held that, with respect to medi-

    cines, it had to be construed strictly and

    narrowly in line with language used in

    the Explanation to 3(d), namely, differ

    significantly in properties with regard to

    efficacy, therefore, that not all advan-

    tageous or beneficial properties are rele-

    vant, but only such properties that di-

    rectly relate to efficacy, which in case of

    medicineis its therapeutic efficacy.

    Thus, holding that the physico-

    chemical properties of the crystalline

    form of imatinib mesylate contended by

    Novartis to be properties with regard

    to efficacy, namely more beneficialflow properties, better thermodynamic

    stability, and lower hygroscopicity, may

    be otherwise beneficial but cannot be

    taken into account for the purpose

    of the test of Section 3(d) of the Act.

    These properties have nothing to do

    with therapeutic efficacy.

    That left bioavailability and its role in

    efficacy. There were two rival contentions

    here. The CPAAargued that in the pharma-

    ceutical field, drug action is explained

    by pharmacokinetics (effect of the body

    on the drug) and pharmacodynamics

    (effect of the drug on the body). Efficacy

    is the capacity of a drug to produce an

    effect, an aspect of pharmacodynamics.

    The generation of response from the

    drug receptor complex is governed by a

    property described as efficacy. Bioavail-

    ability, on the other hand, is a pharma-

    cokinetic property. It is the term used to

    indicate the extent to which a dose of

    drug reaches its site of action or a bio-logical fluid from which the drug has

    access to its site of action.

    Shamnad Basheer, intervenor-cum-

    amicus, argued that safety or significantly

    reduced toxicity should also be taken

    into consideration to judge enhanced

    therapeutic efficacy of a pharmaceutical

    product in terms of Section 3(d).

    While the Court left these submissions

    untouched, it held that bioavailability byitself may or may not result in its efficacy

    being affected. In terms of Section 3(d),

    it must be claimed and established by re-

    search data with in vivo animal models,

    which Novartis had not done. Therefore,

    the claimed invention,crystalline form

    of imatinib mesylate, failed the test of

    Section 3(d). Thus, Novartiss appeal

    came to be dismissed, and the others by

    the CPAAand Natco allowed.

    The Novartis decision has enormous

    significance. It clarifies the meaning of

    efficacy in Section 3(d), stating that mere

    advantages are insufficient. The applicant

    has to show with in vivo animal models

    how therapeutic efficacy is significantly

    enhanced. Crucially, the relentless lobby-

    ing by western pharma MNCsto whittle

    down Section 3(d) has been repelled.

    Notes

    1 The unfinished agenda is now being pursuedthrough the free trade agreements.

    2 These deliberations included the Tek Chandand Ayyangar Committees and various Parlia-mentary Committees.

    3 Earl ier Parliament had refused to introducethis, as a result of which India was taken to theTRIPS Disputes Council where India lost. Indiathen appealed again. It lost again. Parliamenthad no choice but to introduce the interimregime of EMR to avoid trade sanctions.

    4 In 1999 the Patent Act was amended to providepatent protection for foods and medicines. The2002 amendments were made to amend Sec-tion 2(1)(j) relating to invention, add 2(1)(ac)relating to industrial application and 2(1)(ja)relating to inventive step, as also Section 83relating to general principles. In 2005 apart

    from amending Section 3(d), amendmentswere carried to Section 2(1)(ja) and the provisionof pre-grant opposition under Section 25(1). Asa result of all the amendments India has beenable to use TRIPS flexibilities to the maximum.This includes patentability criteria, pre-grant,post-opposition, revocation procedures, counterclaim in infringement suits to set aside the grantof the patent, provisions for compulsory licences,government use, etc.

    5 The World Health Organization (WHO) andthe UNAIDS, amongst others, had appealed toIndian Parliamentarians to keep in mind thatIndia was the pharmacy of the poor in the de-

    veloping world while making the final amend-ment (see Sengupta 2013).

    6 The Doha Declaration of 11 December 2001

    issued by the Inter Ministerial Conference ofthe WTO recognised the gravity of publichealth problems and that the TRIPS Agreementhas to be interpreted to protect public health.

  • 7/24/2019 The Glivec Precedent

    10/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 49

    7 Interestingly the letter from the WHO wassigned by the present president of the WorldBank Jim Young Kim.

    8 Thus, in India the MNCs have, with the localindustry formed the US-India Business Coali-tion, which has issued documents aboutthe need to delete Section 3(d) from thePatents Act.

    9 Under the Indian Patents Act any person can filea pre-grant opposition on the grounds indicated

    in Section 25(1). Post-grant opposition can befiled only by a person interested.

    10 The study was conducted on rats, but the com-parison was with the base form of imatinib and

    not with the mesylate form of imatinib, whichwas also known at that time.

    11 Madras High Court Writ Petition No 24759 and24760 of 2006, at para 19.

    12 Referring to the imatinib base and the mesylatesalt form of imatinib.

    13 Application of John Paul Hogan and Robert LBanks, 1977 (559 F.2d 595).

    14 The Court did go into the issue of novelty ornon-obviousness of the crystalline form imat-

    inib mesylate. It dealt with the issue of the ap-plicability of Section 3(d).

    15 Monsanto Company vs Coramandal IndagProducts (P) Ltd, 1986 (1 SCC 642).

    References

    Correa, Carlos M (2013): Is Section 3(d) Consistentwith TRIPS?,Economic & Political Weekly, 48(32).

    NIHCM (2002): Changing Patterns of Pharmaceuti-cal Innovation(Washington: National Institutefor Health Care Management).

    Sengupta (2013): Two Decades of Struggle,Economic & Political Weekly, 48(32).

    Waning, Brenda, Ellen Diedrichsen and Suerie Moon

    (2010): A Lifeline to Treatment: The Role ofIndian Generic Manufacturers in Supplying Anti-retroviral Medicines to Developing Countries,

    Journal of the International AIDS Society, 13: 35.

  • 7/24/2019 The Glivec Precedent

    11/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 49

    Is Section 3(d) Consistent

    with TRIPS?

    Carlos M Correa

    This article looks at how

    inventions are assessed by various

    policy regimes in the world and

    analyses the Indian regulations

    in this light. A well-formulated

    and scientific legislation to apply

    the inventive step standard could

    avoid most of the complications

    caused by Section 3(d) of theIndian Patents Act, which is

    compatible with the Trade-Related

    Aspects of Intellectual Property

    Rights Agreement, but about

    which questions can still be raised.

    Novartis challenged the consistency

    of Section 3(d) with regard to

    the Agreement on Trade-Related

    Aspects of Intellectual Property Rights

    (TRIPS) before the Madras High Court.

    The high court refused to entertain this

    argument. Interestingly, the Swiss gov-

    ernment did not initiate a case in the

    World Trade Organization (WTO) in sup-

    port of Novartiss argument regarding

    the alleged violation of TRIPS. However,the Supreme Court decision has not

    foreclosed the possibility of such com-

    plaints and an analysis of the subject is

    still relevant.

    On the one hand, the United States

    Trade Representative (USTR) which

    has regularly cited Section 3(d) as

    one of the reasons to keep India on its

    list of countries whose intellectual

    property rights regimes are of concern

    to the US (Sampat et al 2012: 414)1

    stated (USTR2013: 38) that the Indian

    Supreme Court decision

    appears to confirm that Indias law creates a

    special, additional criterion for select tech-

    nologies, like pharmaceuticals, which could

    preclude issuance of a patent even if the ap-

    plicant demonstrates that the invention is

    new, involves an inventive step, and is capa-

    ble of industrial application.

    This statement suggests the possible

    incompatibility of Section 3(d) with two

    aspects of Article 27.1 of the TRIPS

    Agreement: (a) the non-discriminationclause,2and (b) the obligation to grant

    a patent when the three patentability

    criteria specified in that article (novelty,

    inventive step/non-obviousness, industrial

    applicability/utility) are met, without im-

    posing additional substantive conditions.

    On the other hand, other countries,

    such as the Philippines, have adopted

    measures similar to Section 3(d) or apply

    policies that limit the patentability of

    certain categories of pharmaceutical

    products. For instance, in Argentina's 2013

    Trade Policy Review a series of specific

    questions were raised regarding recently

    adopted guidelines on the patentability

    of pharmaceutical products and proc-

    esses3that suggested possible inconsist-

    encies with Article 27.1 of the TRIPS

    Agreement. Some of those questions

    (posed by Japan, the European Union)

    specifically addressed the patentabilityof crystalline forms, the subject matter

    of Novartis s patent application in India.

    Does It Discriminate?

    The obligation not to discriminate con-

    tained in Article 27.1 applies both to the

    availability and enjoyment of patent

    rights, meaning that neither the acquisi-

    tion of patent rights nor the means

    for enforcement can be subject to dis-

    crimination. Although the provision is

    broad in this respect, it is only applica-

    ble when discrimination takes place on

    the basis of one of the following grounds

    (1) the place of invention, (2) the field

    of technology, and (3) whether prod-

    ucts are imported or locally produced

    (Correa 2007).

    Arguments about the possible incon-

    sistency of Section 3(d) with the TRIPS

    Agreement allude to the second ground

    the field of technology.

    A possible conflict with the non-discrimination clause was unsuccessfully

    claimed by the European Commission (EC)

    Carlos M Correa ([email protected]) teaches at

    the University of Buenos Aires and is Advisoron Intellectual Property and Trade, South

    Centre, Geneva.

  • 7/24/2019 The Glivec Precedent

    12/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly50

    in its complaint against Canada relating

    to the Bolar exception (WTO2000).The

    panel rightly made a distinction between

    discrimination and differentiation.4

    Section 3(d) is not discriminatory

    under the terms of Article 27.1 of TRIPS.

    First, it does not apply only to pharma-

    ceutical products, but to any chemicalproduct. It may be applied, for instance, to

    examine the patentability of an isomer of

    an agrochemical product. Second, even if

    (for the sake of argument) Section 3(d)

    applied only to inventions in the pharma-

    ceutical field, it would be justified by the

    need to take the specific characteristics

    of such products into account in the

    examination process. Indeed, special

    considerations regarding the type of

    claims (compositions, salts, polymorphs,

    etc) are unavoidable whether explicitly

    contained or not in laws or other regula-

    tions, to assess the patentability of any

    claimed product in that field. The same

    applies to other technological fields, such

    as biotechnology and computer software.5

    There are, in fact, many examples of

    guidelines and legal provisions that

    specifically address the case of pharma-

    ceutical inventions in national laws and

    policies, including in developed countries.

    Thus, several patent offices have adoptedspecific criteria to examine chemical

    and/or pharmaceutical patents, without

    objection from any WTO member. For

    example, the Japanese Examination

    Guidelines for Patent and Utility Model

    contains a specific chapter on Medicinal

    Inventions (PartVII, Chapter 3).

    Moreover, some national laws contain

    provisions specifically related to phar-

    maceuticals which have never been chal-

    lenged under the WTO rules: the French

    industrial property law provides for the

    grant of compulsory licences on patents

    relating to medicines (Article L613-16);

    the Australian USFree Trade Agreement

    Implementation Bill 20046 introduced

    anAU$10 million penalty for drug patent

    litigation in bad faith; under USlegislation

    (35 USCSection 156) and in accordance

    with the free trade agreements (FTAs)

    signed by the USwith a number of coun-

    tries,7 special provisions apply for the

    extension of the term of pharmaceuticalpatents to compensate for delays in the

    marketing approval of medicines.

    Another potential objection to Sec-

    tion 3(d) under Article 27.1 of the TRIPS

    Agreement, as suggested in the USTR

    2013 Report quoted above, is that it

    imposes an additional standard to obtain

    a pharmaceutical patent, not provided

    for and in violation to the first sentence

    of Article 27.1.There have been different interpreta-

    tions in the context of the Novartis

    case regarding what type of standard

    Section 3(d) actually establishes.

    The Intellectual Property Appellate

    Board (IPAB) competent to hear appeals

    from the decisions of the Indian patent

    office characterised the efficacy test

    imposed by that provision as an enhanced

    inventive step requirement. While the

    IPABconsidered that Novartiss crystalline

    form met the ordinary inventive step

    standard, it argued that it failed to meet

    the stricter inventive step standard

    demanded by Section 3(d).8

    Invention and Patentability

    In the view of the Indian Supreme Court

    the crystalline form of imatinib me-

    sylate fails in both the tests of invention

    and patentability as provided under

    clauses (j), (ja) of Section 2(1) and Sec-

    tion 3(d) respectively (para 195).9It has also been suggested that

    Section 3(d) relates to the utility require-

    ment for patentability: the limited inte-

    gration of efficacy considerations, more

    traditionally seen in drug-marketing laws,

    is a sound and long-overdue attempt to

    rectify the low level of proof of real

    util ity that mars patent regimes (Roderick

    and Pollock 2012).

    Whether Section 3(d) is part of the defi-

    nition of invention (as explicitly stated by

    the provision)10or a patentability require-

    ment (inventive step and/or utility), is not

    essential for an assertion of the provisions

    compatibility with Article 27.1 of the

    TRIPSAgreement. Such compatibility can

    be sustained under both interpretations.

    In effect, an important flexibility that

    WTO members enjoy under the TRIPS

    Agreement is to define what is to be con-

    sidered an invention for the purposes

    of patent law. Like most patent laws in

    the world, the TRIPSAgreement does notdefine what an invention is. Hence, WTO

    members can adopt different concepts,

    in accordance with their legal traditions

    and national policies, as long as this is

    made in good faith and in accordance

    with the interpretative criteria codified

    by the Vienna Convention on the Law of

    the Treaties (Articles 31 and 32).

    As a result of the policy space left by

    the TRIPSAgreement, national laws candistinguish between inventions that

    are patentable and other matters that

    are not, as many laws actually do.11

    The TRIPSAgreement permits diverse

    approaches regarding the interpretation

    of the obligation to patent any inven-

    tions. As noted in a World Intellectual

    Property Organization (WIPO) study, [A]t

    the national/regional level, the exclusions

    from patentable subject matter provided

    for in national/regional legislation vary

    significantly (WIPOSecretariat 2009: 3).

    One example is the different treatment

    conferred under various national regimes

    to substances found in nature such

    as genes.12

    If Section 3(d) were deemed to define

    a patentability requirement (as held by

    the IPAB) then similar considerations

    would apply. While Article 27.1 obliges

    WTOmembers to grant patents where the

    specified standards are met, it does not

    define them. This important flexibilityallowed, for instance, the USto maintain

    until recently and defend the TRIPScom-

    patibility of a novelty standard (35 USC

    Section 102(a)) that combined local and

    universal novelty depending on whether

    the disclosure of the invention had taken

    place within or outside the territory of

    the US. The US held at the Council for

    TRIPSthat in the TRIPSAgreement there

    was no prescription as to how WTO

    members define what inventions are to

    be considered new within their domestic

    systems and, hence, that its legislation

    was perfectly consistent with the provi-

    sions of the TRIPSAgreement.13

    In particular, the TRIPS Agreement

    does not define the standard of non-

    obviousness or inventive step: it does

    not limit the WTO members right to

    choose whether to apply more or less

    rigorous criteria to grant a patent. Al-

    though under most patent laws those

    concepts require an enquiry as t0 whetherthe claimed invention is evident or obvi-

    ous to a person with skills in the relevant

  • 7/24/2019 The Glivec Precedent

    13/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 51

    technical field, there is in fact no uni-

    formity in the way the standard is

    defined and applied.

    Given this important flexibility under

    the TRIPSAgreement, WTOmembers can

    adopt patentability criteria that avoid

    the evergreening of pharmaceutical

    patents, that is, the grant of patents onminor developments whose protection is

    sought to delay or block generic com-

    petition. Preventing evergreening was the

    very purpose of Section 3(d) as extensively

    elaborated on by the Indian Supreme

    Court in the commented decision.14The

    rejected Novartis patent application, as

    noted, referred to a crystalline form or

    polymorph of imatinib mesylate, not

    to the drug itself (imatinib), or its

    particular salt (mesylate).

    Several polymorphs may exist for the

    same chemical compound. A polymorph

    is not actually invented; it is a property

    inherent to such a compound which is

    found in the process of crystallising a

    given substance (depending on solvent,

    heating, stirring, and other conditions)

    or as the result of the transformation

    (without human intervention) of the ar-

    rangements of the molecules in thesolid-

    state structure.15Hence, it may be argued

    that a polymorph is not an invention, butrather the outcome of a discovery.

    If, however, a polymorph would be

    deemed an invention, it would not meet a

    rigorous inventive step/non-obviousness

    standard since, for a person working in

    the pharmaceutical industry, the need

    to try and obtain different polymorphs

    and to choose the most suited for pro-

    duction is obvious. For a person with

    basic knowledge in organic chemistry, it

    is well known, in effect, that one poly-

    morph may be more stable and have

    better properties than others for manu-

    facturing a particular drug (Purohit and

    Venugopalan 2009: 890-91).Hence, even

    if a particular polymorph showed a

    significant enhancement of the drugs

    therapeutic effect, a patent office may

    consider it as not patentable.

    This suggests that the same decision

    to reject a patent application regarding

    a crystalline form may be reached

    even in the absence of a provision likeSection 3(d). The guidelines for the

    examination of pharmaceutical patents,

    published by the International Centre

    for Trade and Sustainable Development

    (ICTSD), World Health Organization

    (WHO), United Nations Conference on

    Trade and Development (UNCTAD) and

    United Nations Development Programme

    (UNDP), for instance, recommended patent

    offices to refuse patent applications onparticular polymorphs (Correa 2006: 11).

    In line with this recommendation, but

    with a more rigorous approach, the Joint

    Resolution adopted by the Argentine

    government in 2012 establishes that, as a

    rule, a new crystalline form of a known

    substance is not patentable.

    This Joint Resolution illustrates that

    an outcome similar to that emerging from

    the Novartis decision can be obtained

    through the application by patent offices,

    in a rigorous manner, of the conventional

    inventive step/non-obviousness require-

    ment. In many instances, patents on

    polymorphs have been held invalid or

    not infringed; even in the US it has

    become more difficult to get a patent

    granted on a polymorph and to defend it

    if challenged in courts (Vure 2011).

    Conclusions

    Section 3(d) is not discriminatory in

    terms of Article 27.1 of the TRIPSAgree-ment. It does creates neither additional

    nor different requirements of patentability

    other than those specified in said article.

    With the adoption of Section 3(d) India

    has made use of the flexibility allowed to

    WTOmembers to determine what is the

    eligible subject matter for the purposes

    of the national patent law. WTO mem-

    bers also have policy space to define the

    specific contours of the patentability

    standards, as the TRIPSAgreement does

    enumerate them, but does not provide

    specific rules governing their definition

    and application.

    Section 3(d) is not only compatible

    with the TRIPSAgreement. As interpreted

    in the Novartis decision commented

    above, it also enshrines the right policy

    approach in dealing with pharmaceutical

    patents: protection should be granted

    when genuine inventions are claimed,

    but rejected when patent applicants just

    aim at creating barriers for generic com-petition through the patenting of a broad

    range of minor, often trivial developments.

    While some countries may be rightly

    encouraged by the outcome of the Novartis

    case to adopt a provision similar to

    Section 3(d) through legislative reform,

    regulations or patent offices guidelines,

    it is worth noting that an equivalent result

    may be obtained through a rigorous and

    scientific application of the inventivestep standard.

    Notes

    1 On occasion of Indias Trade Policy Review(2011) at WTO, the US questioned whether theGovernment of India considered the require-ment under Section 3(d) to be in line withTRIPS Article 27(1). Indias reply indicated thatthe provisions of Section 3(d) of the Patents

    Act, 1970 are fully compliant with Article 27(1)of the TRIPS Agreement read with Article 8 ofthe said Agreement.

    2 Article 27.1: patents shall be available andpatent rights enjoyable without discrimination

    as to the place of invention, the field of techno-logy and whether products are imported orlocally produced.

    3 Joint Resolution of the Ministry of Industry(118/2012), Ministry of Health (546/2012) andInstituto Nacional de la Propiedad Industrial(107/2012).

    4 See WTO (2000), para 92. It is worth notingthat the TRIPS Agreement does differentiatethe treatment given to semiconductor technol-ogies (Article 31(c)). There is also specifictreatment for the textile sector in the area ofindustrial designs (Article 25.2).

    5 For instance, the US patent office and courts ap-ply differently the non-obviousness and disclo-sure standards to biotechnological and softwareinventions (Burk and Lemley 2003).

    6 Amendment (2), The Senate, The Parliament ofthe Commonwealth of Australia (codified as newSection 26C of the Therapeutic Goods Act 1989).

    7 See Singapore FTA, Article 16.8.4(a); Chile FTA,Article 17.10.2(a); Morocco FTA, Article 15.10.3;Bahrain FTA, Article 14.8.6(b)(i); CAFTA,

    Article 15.9.6(b).

    8 See the IPAB decision of 26 June 2009, p 189.

    9 The Court, however, emphasised that in which-ever way Section 3(d) may be viewed, whetheras setting up the standards of patentability oras an extension of the definition of invention,it must be held that on the basis of the materialsbrought before this Court, the subject product,that is, the beta crystalline form of ImatinibMesylate, fails the test of Section 3(d), too, ofthe Act (para 190).

    10 Section 3 (chapeau): What are not inventions.The following are not inventions within themeaning of this Act.

    11 See, for instance, Article 52(2) of the EuropeanPatent Convention.

    12 The controversy regarding the patentability ofhuman genes in the US is illustrative of the

    various views on the matter found even at thenational level (USDJAMP et a l 2010).

    13 See WTO document IP/Q3/USA/1, 1 May 1998.

    14 See, e g, para 100-102.15 For instance, in the case of ritonavir (an anti-

    retroviral), after 18 months of its launch itsmanufacturing company Abbott observed anunexpected occurrence. The final product did notshow dissolution and the drug was precipitating.

    After considerable investigations it was revealedthat this was because of a new thermodynamicallymore stable and less soluble polymorph Form-II(Purohit and Venugopalan 2009: 890-91).

  • 7/24/2019 The Glivec Precedent

    14/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly52

    References

    Burk, D and M Lemley (2003): Is Patent Law Tech-nology-Specific?, 17Berkeley Tech Law Journal,(1155), 1155-1206.

    Correa, C (2006): Guidelines for the Examination ofPharmaceutical Patents: Developing a PublicHealth Perspective(Geneva: WHO, ICTSD andUNCTAD).

    (2007): Trade Related Aspects of Intellectual

    Property Rights (Volume VI of Commentaries onthe GATT/WTO Agreements) (Oxford: OxfordUniversity Press).

    Purohit, R and P Venugopalan (2009): Polymor-phism: An Overview, Resonance , September,882-93, viewed on 8 July 2013, http://www.ias.

    ac.in/resonance/September2009/p882-893.pdfRoderick, P and A Pollock (2012): Indias Patent

    Laws Under Pressure, The Lancet, 380(9846):e2-e4, v iewed on 8 July 2013, http://www.the-lancet.com/journals/lancet/article/PIIS0140-6736(12)61513-X/fulltext

    Sampat, B, K Shadlen and T Amin (2012): Chal-lenges to Indias Pharmaceutical Patent Laws,Science, (337): 414-15.

    USTR (2013): 2013 Special 301 Report, United

    States Trade Representative, viewed on 8 July2013, http://www.ustr.gov/sites/default/files/05012013%202013%20Special%20301%20Re-port.pdf

    USDJAMP et al (2010): Brief for the United Statesas Amicus Curiae in Support of Neither Party,

    submitted by the US Department of Justice inAssociation for Molecular Pathology, Petitionersvs Myriad Genetics, Inc et al, viewed on 8 July2013, http://graphics8.nytimes.com/packages/pdf/business/genepatents-USamicusbrief.pdf

    Vure, P (2011): Polymorph Patents: How StrongThey Are Really?, International Journal of

    Intellectual Property Management, 4(4): 297-306.

    WIPO Secretariat (2009): Exclusions from PatentableSubject Matter and Exceptions and Limitations

    to the Rights, SCP/13/3,F, viewed on 8 July 2013,http://www.wipo.int/edocs/mdocs/scp/en/scp_13/scp_13_3.pdf

    WTO (2000): Canada Patent Protection for Phar-maceutical Products, Report of the WTO Panel,WT/DS114/R.

  • 7/24/2019 The Glivec Precedent

    15/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly52

    Drop the CaseCampaigning against Novartis

    Leena Menghaney

    Evergreening and the issues this

    practice of abusive patenting

    raised were first highlighted

    in the late 1990s with the HIV

    epidemic, when medicines were

    priced way out of reach. The

    Supreme Courts rejection of

    Novartiss patent has set in placean important safeguard to ensure

    production of affordable generic

    medicines from India.

    That patents on medicines can

    make the difference between life

    and death for millions, first hit

    home with the AIDS pandemic, when

    medicines to treat HIVwere priced way

    out of reach of patients and governments

    in developing countries.1In 1999, in the

    wake of Mdecins Sans Frontires (MSF)

    being awarded the Nobel Peace Prize,

    MSFlaunched the Access Campaign. At thetime patented HIV treatment cost more

    than $10,000 per patient per year. One

    of the first tasks of the Access Campaign

    was to challenge the high costs of exist-

    ing drugs such as those to treat HIV/

    AIDS and working to bring prices down

    for its medical projects in South Africa

    and Thailand.

    Evergreening and AIDS Treatment

    Evergreening and the issues this practice

    of abusive patenting raised were first

    highlighted in the late 1990s, when the

    HIVepidemic was devastating communi-

    ties particularly those in Africa and

    the barriers to providing treatment

    seemed insurmountable.2

    Zidovudine (AZT) (previously Azidothy-

    midine), one of the first antiretrovirals

    (ARVs) to treat HIV, was a known cancer

    product,3but GlaxoSmithKline (GSK) was

    granted a patent for its use to treat HIV,4

    making it one of the first examples ofan evergreened drug used to treat HIV.

    Similarly, GSK filed a secondary patent

    in 1997 on a formulation containing

    lamivudine andAZT, which was granted

    in several countries, including South

    Africa.5 Access to these ARVs then be-

    came difficult and expensive because of

    these patents.

    The answer to a sustainable supply of

    affordable HIVmedicines lay in India,

    where policies that fostered production

    of and access to affordable generics had

    taken hold in the absence of product

    patents (Sengupta 2013).6The countrys

    generic producers took up the challenge

    in 2001 to develop genericARVs, which

    they sold for just $1 per day, a fraction of

    the price that multinational pharma-

    ceutical companies were then charging.

    With Indian producers able to manu-facture and export fixed dose combina-

    tions ofARVs, the World Health Organi-

    zation (WHO) launched the Treat 3 Million

    by 2005 initiative in 2003. The same year,

    an HIVtreatment programme was set up

    in South Africa the country with the

    highest burden of HIV/AIDS7 through

    the public health system, with India

    following in 2004.

    Indias Patents Act: What Future?

    In 2005, as the deadline for India to fully

    implement the Trade-Related Aspects

    of Intellectual Property Rights (TRIPS)

    Agreement8approached, the country was

    obliged to introduce product patents for

    medicines. Until then, generic producers

    had supplied medicines without fear of

    facing protracted and expensive patent

    infringement suits. Thousands of patent

    applications on medicines had been

    filed since 1995 in anticipation of the

    amendments in Indias patent regime,and a large number including those

    for HIV, tuberculosis (TB) and cancer

    Leena Menghaney ([email protected]) is Campaign Coordinator (India) for the

    Mdecines San Frontires Access Campaign.

  • 7/24/2019 The Glivec Precedent

    16/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 53

    could be considered as evergreening

    patent claims.

    As a result, a single medicine had many

    secondary patent applications pending

    in the Indian patent office covering one

    or more specific features, including

    formulation (e g, heat stable tablets

    and capsules, syrups, etc), combinations(e g, a fixed-dose combination when dif-

    ferent HIV medicines are combined in

    the same pill), and derivatives (salts,

    prodrugs, crystals and polymorphs).

    Concerned about the impact on access

    to medicines, MSF, UNAIDS, WHO and

    even the French Prime Minister Jacques

    Chirac, stepped in to warn the Indian

    government to find the right balance

    between protecting production and access

    to affordable generics, and coming into

    compliance with TRIPS.

    In the midst of civil society protests,

    big pharma lobbying, and international

    media attention, Parliament passed the

    new Patents Act in March 2005 to allow

    for pharmaceutical product patents

    something the country had not done

    since 1970. Although not perfect,9 the

    law crucially allowed for any party to

    oppose a patent before it is granted (Sec-

    tion 25), and the scope of patentability

    under Section 3(d) was restricted toavoid evergreening, the practice of

    granting multiple secondary patents on

    an existing medicine.

    Novartis vs Union of India

    A Novartis patent application on the

    anti-cancer drug imatinib mesylate10

    was one of the first to be examined,

    opposed and rejected by an Indian

    patent office on grounds that it claims

    covered a new form of a known sub-

    stance Section 3(d) of the new law

    and lack of an inventive step. Angered

    by this decision, Novartis went to court

    in May 2006 alleging, among other

    things, that Indias Section 3(d) was

    contrary to the TRIPS Agreement and

    was unconstitutional.

    The patent rejection order obtained

    after Cancer Patients Aid Association

    and others filed a pre-grant opposition

    set an important precedent for the exa-

    mination of patent applications on HIVmedicines. More importantly, Section 3(d)

    had already demonstrated its importance

    in upholding the rejection of the patent

    on imatinib.

    If Novartis succeeded in weakening

    the interpretation of Section 3(d) in

    order to obtain a patent on imatinib

    mesylate, this would defeat its purpose

    in forestalling the practice of evergreen-

    ing. The Indian Patent Office would haveto apply the same standards of patent-

    ability on existing patent applications

    covering HIVmedicines and their differ-

    ent forms, meaning HIVmedicines could

    be easily patented and generic compa-

    nies could face expensive infringement

    suits. This could ultimately result in hav-

    ing a chilling effect on the availability of

    affordable generic medicines.

    The Campaign

    In December 2006, MSFs Access Cam-

    paign, in partnership with Oxfam and

    Delhi Network of Positive People (DNP+),

    launched a petition appealing to people

    to urge Novartis to Drop The Case in

    order to protect the pharmacy of the

    developing world (Doctors Without

    Borders 2006).

    The campaigners faced the daunting

    task of educating ordinary people about

    access to medicines, pharmaceutical

    patents and the impact of the case on theseissues. The message needed to be simple

    this case was a direct attack on the safe-

    guards in Indias patent law and risked

    dismantling Indias capacity to produce

    quality, affordable generic medicines.

    The launch of the campaign turned

    what otherwise promised to be a complex

    patent dispute followed by just a few

    technical organisations and patient groups

    into a global outcry against the greedy

    practices of pharmaceutical companies.

    The petition received a groundswell of

    support from MSF field workers, HIV

    organisations, and ordinary people from

    around the world, resulting in close to

    half a million signatures by August 2007

    when Novartis lost its case at the Madras

    High Court fuelled by protests before

    Novartis s offices worldwide.

    In the wake of two failed appeals,

    Novartis took its final bid for a patent

    and now a challenge on the interpreta-

    tion and application of Section 3(d) tothe countrys Supreme Court. If this

    challenge were successful, it could take

    much of the substance out of the public

    health provision, allowing evergreen-

    ing, and, as Correas (2013) accompany-

    ing article explains, narrowing the flexi-

    bility in TRIPS.

    MSF relaunched the Drop The Case

    campaign, this time rebranded as Stop

    Novartis, in February 2012. While theinitial campaign took the form of a peti-

    tion, the Stop Novartis campaign took

    advantage of the global reach of Twitter.

    The social media platform was used to ask

    people to target the company directly,

    through the companys @Novartis pro-

    file and the hashtag of #STOPNovartis.

    The new campaign almost immediately

    goaded Novartis into responding, with a

    series of tweets defending the companys

    record and documents that attempted to

    set the record straight on its actions

    from its own perspective.

    Novartis: What They Had to Say

    Over a period of seven years, civil society

    and MSF received thousands of media

    queries from journalists who had picked

    up on the campaign. Public statements

    from Novartis covered the most com-

    mon misconceptions on the subject of

    pharmaceutical patents.

    These included Novartiss statementthat as imatinib mesylate had been

    granted a patent in 40 countries, includ-

    ing Switzerland and the US, India should

    simply follow suit. In using this tactic,

    Novartis conveniently glossed over the

    fact that countries have at their disposal

    TRIPS flexibilities to adopt different

    standards in designing their patent sys-

    tems to best suit their own needs a

    point well elaborated in Correa (2013).

    Accordingly, it is crucial that developing

    countries, in particular, make a careful

    decision in what should be allowed a

    patent and what should not.

    MSF was clear in its response: India

    must stand firm, applying a patentability

    standard that weeded out evergreened

    patent claims. Its generic production of

    medicines saved millions of lives.

    Novartis also tried predictably to

    justify its patent claims in India by

    stating its research and development

    (R&D) costs needed to be recoupedthrough the drugs high cost (Ghose 2013).

    The company which claims patent

  • 7/24/2019 The Glivec Precedent

    17/20

    THE GLIVEC PRECEDENT

    august 10, 2013 vol xlv ii i no 32 EPW Economic & PoliticalWeekly54

    rights in 40 countries, reaping huge

    profits in return deliberately under-

    played the significant contribution made

    by the public sector and philanthropic

    sources in the US11 meaning it actually

    spent just a fraction of the total R&Dcost

    (Love 2013).

    Throughout the court challenges andin the wake of the Supreme Courts

    decision, which Grovers (2013) article

    discusses, Novartis was quick to raise

    fears about the future of innovation in

    India if patents were rejected by the

    patent office (Novartis 2013). This was a

    lazy argument, in that the case was not

    about India refusing to grant patents on

    new medicines, but stemmed from

    whether a salt form of an existing drug

    deserved a patent in India. It further

    opened the debate on the current sys-

    tem, that despite patent protection, the

    diseases and health issues of developing

    countries are neglected by companies

    (Menghaney 2013).

    What Worked?

    The Supreme Court decision on 1 April

    was the final act in a legal battle that

    stretched back to 2004. The verdict,

    when read with the law, sets a high

    standard for the various tests in the pat-ent legislation, particularly the efficacy

    test of Section 3(d) and the inventive

    step requirement. In the future, this will

    lead to fewer patents on new forms of

    known medicines, an important safe-

    guard to ensure production of affordable

    generic medicines from India.

    While the campaign ultimately failed

    to convince Novartis to Drop The Case, it

    effectively forged alliances in civil society

    in creating an international debate on

    the abusive practice of evergreening pat-

    ents. This groundswell of support and

    the local and international outcry on the

    daring tactics pharmaceutical companies

    will stoop to in order to secure profit, set

    the tone of the campaign and formed the

    backdrop to the legal case.

    Ultimately, the campaign showed that

    there is no reason for developing countries

    to have a system that blindly hands out

    patents when clear measures to protect

    peoples access to medicines and preventabusive patenting practices by the pharma-

    ceutical industry can be taken.

    Notes

    1 In March 2000, the daily dose (400 mg) of flu-conazole needed to treat cryptococcal menin-gitis cost $17.84, more than two t imes the daily

    wage of an average employed South Africanwho earned just $7.69. In Thai land, however,the daily dose of fluconazole costs just $1.20(Doctors Without Borders 2000).

    2 A common patenting practice in the pharma-

    ceutical industry aimed at filing and thenobtaining separate (and sequential) patents re-lating to different aspects of the same medi-cine, such as different dosages, formulations,fixed dose combinations and different forms ofthe active ingredient, among others.

    3 In September 1986, early clinical tests showedthat Azidothymidine (AZT), a drug first syn-thesised in 1964 to be used as chemotherapyfor leukemia, slowed down the progress of thedisease. In 1987, AZT became the first anti-HIVdrug to be approved by the US FDA.

    4 US4724232, Glaxo Wellcome, February 1988,viewed on 30 June 2013, http://patft.uspto.gov/netacgi/nph-Parser?Sect1=PTO1&Sect2=HITOFF&d=PALL&p=1&u=%2Fnetahtml%2FPTO%2Fsrchnum.htm&r= 1&f=G&l= 50& s1=

    47 24 32.PN. &OS=PN/ 4724232& RS= PN/4724232

    5 ZA9709726, Glaxo Wellcome, 1997, viewed on30 June 2013, http://apps.who.int/medicine-docs/en/d/Js4913e/7.html

    6 Under the 1970 Patents Act, India did not grantproduct patents on medicines.

    7 On 19 November 2003, the South AfricanNational Department of Health announced anOperational Plan for the rollout of ARV medi-cines in the public health system. UNAIDS/WHO statistics from 2004 suggested that of theapproximately 5.3 million people living withHIV/AIDS in the country, 7,50,000 were inneed of ARV treatment.

    8 The TRIPS Agreement came into effect on 1

    January 1995, setting out minimum standardsfor the protection of intellectual property,including patents on pharmaceuticals. Underthat agreement, since 2005 new drugs may besubject to at least 20 years of patent protect ionin all, apart from in the least-developed coun-tries and a few non-World Trade Organizationmembers, such as Somalia.

    9 See Gopakumars (2013) accompanying article.

    10 Marketed as Glivec.

    11 See Dutfields (2013) article for a discussion.

    References

    Correa, Carlos M (2013): Is Section 3(d) Consist-ent with TRIPS?,Economic & Political Weekly,48(32).

    Doctors Without Borders (2000): One World, OnePrice Means Death for People With AIDS inPoor Countries, 13 March, viewed on 30 June2013, http://www.doctorswithoutborders.org/press/release.cfm?id=536

    (2006): MSF Urges Novartis to Drop CaseAgainst Indian Government, 20 December,viewed on 30 June 2013, http://www.doctor-swithoutborders.org/press/release.cfm?id=3715&cat=press-release

    Dutfield, Graham (2013): Who Invented Glivec?Does It Matter Anyway?,Economic & Polit icalWeekly, 48(32).

    Ghose, Sagarika (2013): FTN: SC Ruling onNovartis: Should Cancer Drugs be Cheaply

    Available?, IBN Live, 2 April, viewed on

    30 June 2013, http://ibnlive.in.com/videos/382785/ftn-sc-ruling-on-novartis-should-can-cer-drugs-be-cheaply-available.html

    Grover, Anand (2013): Analysing the SupremeCourt Judgment,Economic & Political Weekly,48(32).

    Love, James (2013): R&D Costs for Gleevec,Knowledge Ecology International, 3 April,

    viewed on 30 June 2013, http://lists.keionline.org/pipermail/ip-health_lists.keionline.org/2013-April/003005.html

    Novartis (2013): Supreme Court Denial of GlivecPatent Clarifies Limited Intellectual PropertyProtection and Discourages Future Innovationin India, 1 April, viewed on 30 June 2013,http://www.novartis.com/newsroom/media-releases/1689290.shtml

    Menghaney, L (2013): R&D: What Novartis Says...and Why Its Wrong,Livemint, 12 April, viewedon 30 June 2013, http://www.livemint.com/Opinion/ 3KaBPvulUVkRSoDh8xkVRJ/RD-What-Novartis-says-and-why-its-wrong.html

    Sengupta, Amit (2013): Two Decades of Struggle,Economic & Political Weekly, 48(32).

    SurveyAugust 27, 2011

    Experimental Economics: A Survey

    by

    Sujoy Chakravarty, Daniel Friedman, Gautam Gupta, Neeraj Hatekar, Santanu Mitra, Shyam Sunder

    Over the past few decades, experimental methods have given economists access to new sources

    of data and enlarged the set of economic propositions that can be validated. This field has

    grown exponentially in the past few decades, but is still relatively new to the average Indian

    academic. The objective of this survey is to familiarise the Indian audience with some aspects

    of experimental economics.

    For copies write to:

    Circulation Manager,

    Economic and Political Weekly,

    320-321, A to Z Industrial Estate,

    Ganpatrao Kadam Marg, Lower Parel,

    Mumbai 400 013.email: [email protected]

  • 7/24/2019 The Glivec Precedent

    18/20

    THE GLIVEC PRECEDENT

    Economic & PoliticalWeekly EPW august 10, 2013 vol xlviii no 32 55

    The Need to Curb Patentson Known Substances

    K M Gopakumar

    Despite a progressive judgment

    by the Supreme Court of India

    on Section 3(d) of the Indian

    Patents Act, the processes of

    legislation and implementation

    are not equipped to uphold the

    spirit of the Novartis judgment.

    This article explains the various

    loopholes that plague the system

    of patents in India, and suggests

    possible solutions.

    Legally speaking, the most impor-

    tant implication of the judgment is

    that the Supreme Court has bro-

    ught a great degree of clarity with re-

    gard to the interpretatio