Sie sind auf Seite 1von 7

Annals of Oncology 22 (Supplement 1): i60i66, 2011

doi:10.1093/annonc/mdq668

symposium article
Social and ethical implications of BRCA testing
A. Surbone*
New York University Medical School, New York, USA

symposium
article

introduction
Breast cancer is highly prevalent and is the second cause of
cancer death in women. In the USA, 180 000 American
women were diagnosed with invasive breast cancer in 2007 and
another 62 000 with in situ or non-invasive breast cancer, while
>40 000 women were expected to die from breast cancer in the
same year [1]. In 2007, 22 500 women were diagnosed with
ovarian cancer, accounting for 3% of all cancers among
women, and 15 280 women were projected to die from ovarian
cancer [1]. Only 10% of all breast cancer is hereditary, and <1%
of the general population is estimated to carry a mutation in
BRCA1 or BRCA2. BRCA mutations are associated with
a relative increased lifetime risk of breast cancer of 2.76.4
times, and a relative lifetime risk of ovarian cancer of 9.335.3
times greater than average risk women [2, 3].
Women who carry a BRCA mutation are given options of
early and intensive surveillance, chemoprevention and
prophylactic surgery. Oncologists and geneticists help women
understand the risk their BRCA status represents for them and
their relatives. They also identify and address biopsychosocial
factors that may foster womens adherence to preventive and/or
risk-reducing strategies, in order to reduce the morbidity and
mortality associated with hereditary or familial cancers [4, 5].
Available risk reduction options in the past 15 years seem to

*Correspondence to: Antonella Surbone, Department of Medicine, Division of Medical


Oncology, New York University Medical School, 550 First Avenue, BCD 556, New York,
NY, USA. E-mail: antonella.surbone@nyumc.org

have significantly lowered the chance that high-risk women


develop the breast and ovarian cancers [6, 7].
Since the initial application of BRCA testing in research first,
and subsequently also in oncology practices, scientists,
physicians and bioethicists have consistently cared about the
medical and psychosocial well-being of mutation carriers, and
have cautioned the public about the limited predictive power of
genetic testing, especially outside high-risk families, due to the
relatively low gene penetrance, the possibility of new mutations
with different significance that are still to be identified, and the
role of environmental factors in cancerogenesis and tumor
progression. Furthermore, preventive and interventional
measures are still being developed and genetic testing carries
potentially negative psychosocial repercussions for individual
carriers and their families [8, 9].
The identification of DNA mutations predisposing to cancer
susceptibility is now expanding to include whole-genome
profiling for personalized approaches to cancer patients and
direct-to-consumer (DTC) genetic testing. Knowledge about
the ethical and juridical implications of genetic testing is
becoming essential for oncologists, who are being asked with
increasing frequency to counsel their patients with respect to
the medical, psychological and social repercussions of genetic
information, even when obtained outside the context of an
established patientdoctor relationship. Many articles and
reviews have addressed the main ethical and social implications
of BRCA testing [1018], including informed consent, privacy
and confidentiality, a persons right to know or not to know
their genetic information, carriers responsibility to share

The Author 2011. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
All rights reserved. For permissions, please email: journals.permissions@oup.com

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

Oncologists are asked with increasing frequency to counsel their patients with respect to the medical, psychological
and social repercussions of genetic testing for cancer susceptibility that may have been prescribed by physicians or
carried out through direct-to-consumer tests. This article critically reviews the main ethical and social implications of
BRCA testing, focusing on genetic responsibility and genetic discrimination. Genetic responsibility toward oneself and
others is a highly debated implication of genetic testing for cancer predisposition that requires broad considerations of
the boundaries between individual and community rights and a reappraisal of the notion of autonomy as relational.
Physicians duty to warn at risk relatives can be an ethical quandary, yet confidentiality is key to the patientdoctor
relationship. Mutation carriers may be subject to different forms and degrees of genetic discrimination and many
individuals at risk have forgone BRCA testing to avoid potential discrimination. The scientific and medical community,
together with patients and the public, has actively engaged against genetic discrimination. The legislation in many
countries now protects against genetic discrimination by insurance companies and employers. Legal and regulatory
issues are not the final answer to discrimination and profound cultural changes are required to create understanding
and acceptance of all differences.

Annals of Oncology

genetic information with relatives at risk, physicians duty to


warn relatives about familial risk, reproductive choices based
on preimplantation diagnosis, appropriate testing of children
and adolescents, equitable access to testing, disposition of
biological samples, and genetic discrimination. This article
focuses on issues of genetic responsibility and genetic
discrimination, which are now being revisited as the availability
of DTC genetic testing rapidly increases [19, 20].

is genetic information different from


other medical information?

genetic discrimination and legislative


efforts
Genetic discrimination against asymptomatic individuals or
their relatives on the basis of actual or presumed genetic
differences or characteristics has been a concern among women
with strong family histories of breast or ovarian cancer and
their oncologists, since genetic discrimination has occurred in
the past for non-oncologic diseases [2326]. BRCA mutation
carriers may also be subject to overt discrimination or to more
subtle forms of estrangement and rejection by their families or
communities, as being regarded as less-than-ideal partners or
parents [10, 15]. Many high-risk women refrain from being
tested, due to fear of discrimination by health insurance or by
actual or potential employers, or fearing discrimination against
their children [27]. Similar concerns have also been reported
among genetic experts and oncologists [28].

Volume 22 | Supplement 1 | January 2011

People who refrain from being tested out of fears of


discrimination from insurers or employers may suffer damage
to their health due to forgoing preventative care measures,
monitoring, and/or other interventions that might follow from
testing. Health care delivery has been affected by fear of genetic
discrimination, as patients have sometimes paid out of pocket,
or used false names, or may have asked their doctors to omit
genetic information from their medical records. The threat of
genetic discrimination may have also hindered medical research
aimed at developing personalized medicine and new treatment
modalities for genetic diseases [29, 30].
Although documenting the exact magnitude of genetic
discrimination is an arduous task, cases have been reported in
the USA with regard to life and health insurance, the
employment market, and in access to higher education and
adoption [23, 26, 28, 31]. While some authors have pointed out
that societys concern for genetic discrimination may have been
excessive, the active engagement of medical and patient
organizations has positively influenced the recent development
of national and international efforts by legislators to regulate
the collection and use of genetic information and the
enactment of anti-discrimination legislation in the USA and
other countries [3234].
Various countries now have legislation and regulations to
protect against discrimination by insurance companies and in
the workplace. Belgium passed a genetic non-discrimination
law in 1990; other European nations followed suit [35, 36]. The
Biomedicine Convention of the Council of Europe and
UNESCO based their legislative efforts on considering
individual patients as the only subject of genetic information
and on explicitly recognizing the right not to know [35].
In 2008, the US Congress passed the Genetic Information
Nondiscrimination Act (GINA) to protect individuals against
discrimination by health insurers and employers on the basis of
genetic information [3742]. GINA prohibits insurance
companies from taking into account genetic conditions or
family history when determining risk assessment, thus
protecting mutation carriers from paying higher insurance
premiums or being denied health coverage. GINA also
prohibits employers from making employment and
promotional decisions based on genetic information, imposing
fines of up to $300 000 per violation.
GINA is, in large part, the result of the anticipatory work of
researchers and clinicians involved in the development of the
Human Genome Project, who raised concerns about the risk of
genetic discrimination at the outset of the project [38]. Among
common genetic tests protected by GINA are tests for BRCA1
and BRCA2 mutations and HNPCC mutations that predispose
to breast, ovarian, colon and other cancers, as well as tests that
help classify the genetic profile of an existing cancer. GINA thus
protects mutation carriers and facilitates research on the
biology and treatment of cancers associated with specific
genetic mutations [37, 38].
GINA does not regulate insurance underwriting based on
a persons current health status, does not mandate coverage for
any particular test or treatment, does not cover life, disability or
long-term care insurance and does not interfere with health
professionals recommendations for genetic testing to their
patients [37]. GINA does, however, substantially lower patients

doi:10.1093/annonc/mdq668 | i61

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

Genetic information refers to genetic testing for patients and/or


for family members up to fourth-degree relatives. A genetic test
is any analysis to detect genotypes, genetic mutations or
chromosomal changes, not including analysis of proteins or
metabolites directly related to a manifested disease. Genetic
information also refers to any manifestation of disease or
disorder in a family member, and/or to the participation of
a person or family member in research that involves genetic
testing, counseling or education. Genetic information thus has
unique aspects with respect to other medical information, as it
carries potential value, but also danger, for individuals other
than the person tested [21, 22].
This and other unique aspects of genetic knowledge have led
to genetic exceptionalism, the separate treatment of the ethical
and legal implications of genetic testing. Genetic exceptionalism
is a dangerous ethical position that can lead to segregation of
a group of persons at risk [22]. The application of the ethical
principles and methodologies used to analyze and resolve ethical
quandaries in medicine in general can be applied to the complex
questions of autonomy, privacy and responsibility raised by
genetic knowledge, as well as to the potential legal conflicts
surrounding genetic testing [8, 10, 14, 22].
Beyond the ethical and legal implications of genetic testing,
experts and the public have expressed different views on the
value of BRCA testing, from stressing the importance of genetic
knowledge for high-risk women as a means to enhance control
of their lives, to worrying about the potentially negative
repercussions of genetic information [10, 22].

symposium article

symposium article

DTC genetic testing: reviving the


ethical debate.
Cancer risk genomic profiling and DTC genetic testing raise
many of the same ethical concerns that were already addressed
above with regard to testing for genes and other tests of genetic
cancer predisposition. Additional concerns in DTC genetic
testing involve the major impact of economic factors that drive
individuals to be tested without proper understanding of the
meaning of genetic testing and its potential repercussions on
them and their family members [19, 20, 5153]. In addition,
the long-term psychological ramifications of genetic knowledge
not accompanied by proper counseling and medical follow-up
are still unknown, and patients privacy and confidentiality
about their genetic information will be difficult to safeguard
against potential exploitation by individuals and corporations.
Even with genetic non-discrimination legislation, mutation
carriers may find themselves ineligible for life insurance and/or
other long-term agreements. Other ethical and social concerns
regard the additional health-care costs associated with carrying
out the tests and dealing with the consequences of their
findings [19, 20, 5053].

i62 | Surbone

According to the American College of Medical Genetics,


genetic testing should be provided to the public only through
the services of an appropriately qualified health-care
professional, who should be responsible for both ordering and
interpreting the test, as well as for pre- and post-test counseling
and follow-up, as needed [19]. Thus, while individuals have
rights to undergo genetic testing, they should be fully informed
of its inherent risks and entitled to the confidentiality of the
testing and its results [5355]. Unfortunately, DTC genetic
testing is not accompanied by proper counseling, and
advertisements for DTC genetic testing are not subject to the
same degree of federal oversight as those for prescription drugs,
thus increasing the possibility of misleading the public and
physicians in the interest of profit [55].
DTC advertisements for genetic testing, based on the obvious
public fear of cancer, seem highly inappropriate, given the
publics limited understanding of genetics and the lack of
comprehensive oversight of the educational value of
advertisements regarding genetic testing [56]. A campaign to
increase awareness among consumers and physicians about
inherited susceptibility for breast and ovarian cancer, and about
the availability of genetic testing for these conditions could be
beneficial. As a consequence of insufficient clinician and/or
patient knowledge or understanding, in fact, inappropriate
referrals and misinterpretation of test results may occur, as well
as inappropriate medical decision making for the patient and
the entire family, including unnecessary health-care
expenditure, unneeded prophylactic surgery, or false
reassurance and less surveillance in women who are found not
to carry a BRCA mutation. Even well-meaning health-care
providers without specific training in genetics or genetic
counseling may also be manipulated into ordering genetic
testing or making inappropriate referrals [56].
DTC genetic testing may also include still unproven genetic
markers for common multifactorial diseases, such as breast
cancer. An important ethical issue is that some techniques may
identify, not diagnostic genetic variants, but surrogate genetic
markers, known as single-nucleotide polymorphisms (SNPs),
which are associated with a very small increased risk of disease
[57]. With regard to breast cancer, for example, the tests that can
be used to identify SNPs in the FGFR2 gene have been associated
with only a 1.2-fold increase in the risk of developing breast
cancer [52]. Among other pressing ethical questions are: who
should have their genome sequenced? who should prescribe the
test? who should provide pre- and post-test counseling? who
should have access to an individuals genetic information? and
what liability may physicians incur especially when treating more
members of one family? [20, 58]. Ensuring proper continuing
education for health-care professionals and implementing public
education policies are needed, along with standards for accurate
marketing of DTC genetic testing [20, 5860].

individual genetic responsibility and


professionals duty to warn: where do
we stand?
Two of the most controversial ethical issues surrounding
genetic testing for cancer predisposition are: (i) whether or not

Volume 22 | Supplement 1 | January 2011

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

risk of genetic discrimination. As of writing, one claim of


genetic discrimination has been filed since GINA was approved,
by a BRCA-positive woman who allegedly lost her
position at work after undergoing bilateral prophylactic
mastectomy [43].
Genetic non-discrimination legislation is based on the fact
that individuals have no control over their genes. While also
persons with low or unknown genetic risk share the increased
health-care cost for carriers of genetic mutations, such
legislation may lead to decreased overall long-term health-care
costs, if prevention or early interventions prove to be costeffective. Given the lack of a clear definition of genetic test and
genetic data in the laws, however, and of a clear distinction
between genetic and non-genetic data in insurance practices,
the efficacy of genetic non-discrimination laws may not be as
complete as anticipated [44, 45]. It has been reported, for
example, that in European countries the enactment of genetic
non-discrimination legislation may have been less effective than
expected in regard to insurance practice, due to lack of clear
definitions of terms and boundaries between genetic and nongenetic tests and data, thus providing only the illusion of
protection [36]. In addition, legislation may unintentionally
have fostered increased discrimination against patients whose
illnesses are related to lifestyle risks that are assumed to be
under the control of the individual [7, 46]. As the interaction
of genetic and environmental factors is not yet fully
understood for many pathologic entities, increasing the
burden of personal responsibility for high-risk takers may
represent a shifting of discriminatory attitudes from one
group of potential patients to another [36]. The ultimate
response to discrimination involves deep cultural changes,
beyond legal and regulatory issues, to foster a nondiscriminatory approach to all illnesses and disabilities [26,
4749]. As Billings wrote in 2008, antidiscrimination laws are
only effective as guideposts to better understanding and
tolerance [50].

Annals of Oncology

Annals of Oncology

Volume 22 | Supplement 1 | January 2011

about whether relatives would wish to be informed [72, 7476].


Personal or gender differences in communication preferences
and styles can contribute to patients initial decisions not to
share genetic information with others, and cross-cultural
differences in ethical norms and legal requirements may
influence public attitudes and decisions to disclose [7681]. At
the individual level, personal context, perception of personal
risk, perceived or actual vulnerability and receptivity of
relatives, content and timing of communication and personal
feelings all shape individual response to whether and when to
share genetic information with at-risk relatives. At the familial
level, proximity of relationship to ones relatives, family
structure and interpersonal dynamics and prior familial
experiences with cancer account for great variability in
disclosure preference. At the community level, cultural context
and roles, as well as gender and age, play an important role in
communication patterns [76].
As progress is being made in our ability to prevent or
ameliorate with medical or life-style interventions the many
pathologic conditions that can be revealed through genetic
testing, the number of relatives who would wish to be informed
is growing in many countries. Physicians thus increasingly face
the challenge of making judgments about when it may be
appropriate to breach confidentiality on the basis of knowledge
about risks and risk-reduction measures [82]. Health
professionals will also require training and support in
discussing genetic risks with their patients and the importance
of sharing results with at-risk family members, as well as in
responding to relatives questions with respect for the
complexity of intrafamilial communication dynamics [76].
In the UK, the General Medical Council has issued guidelines
urging physicians to judge whether their duty to make the care
of the patients their first concern is greater than their duty to
help protect the other person from serious harm [58, 83]. This
quandary may be exacerbated when physicians are caring for
different members of the same family. Most international
medical organizations and professional ethics codes support the
notion that health professionals obligation is toward the
patient, unless there is a risk of serious harm to others [8, 35,
62, 65]. If interventions could prevent potentially serious
medical conditions, however, physicians may have to consider
overriding confidentiality, and must also reflect on their
potential future liability in regard to those family members
[58]. It has been suggested that certain genetic information may
be seen not only as the result of an individuals test subject to
full confidentiality, but also as part of a familial joint account
open to being used in guiding treatment or testing for the
benefits of other family members, even when not meeting the
seriousness criterion [84]. The recent presentation and
discussion of two clinical cases by Parker and Lucassen
powerfully illustrates a contemporary diagnostic dilemma, in
which the gene penetrance and the interaction of genetic and
environmental factors on the one hand, and the magnitude of
risk reduction with available interventions on the other, are
difficult to assess in many cases of positive genetic testing [58].
Difficult decision making on how to manage genetic
information in patients and their relatives is bound to increase
for all physicians and for clinical oncologists in particular. The
first and essential step is to explain to patients the nature of the

doi:10.1093/annonc/mdq668 | i63

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

individuals have a responsibility to acquire genetic knowledge


and to share positive results with family or community
members at risk [61]; (ii) whether or not health professionals
have a responsibility to disclose relevant genetic information to
their patients relatives; and (iii) how they should best counsel
those mutation carriers who may decide not to share genetic
information [6265].
Genetic responsibility toward oneself and others is a highly
debated implication of genetic testing for cancer predisposition.
Genetic links in high-risk families would appear to be
associated with moral obligations that go beyond those related
to individual autonomy [61, 66]. In addition, the questions of
whether and when to share genetic information requires broad
consideration of the boundaries between individual and
community rights and a reappraisal of the notion of autonomy
as relational [66, 67]. Understanding autonomy in a relational
way entails recognizing the internal and external factors that
shape our decision making and the responsibilities that arise
from our connections to others, as well as acknowledging the
cultural, socio-economic and contextual aspects of patients
decision making [6668]. Yet it seems medically premature to
consider genetic testing as a necessary step for people to take in
regard to their future health status [69]. In addition, the
psychological and social ramifications of genetic testing are so
complex that questions about whether and when to be tested
are legitimate ones [69].
In the ethical and medical literature, there is general
agreement that genetic information, by virtue of its potential
repercussions on the health of an entire family or community,
reminds us of the ties we have with each other. Thus there is
a compelling moral argument for affirming the importance of
sharing genetic information with relatives who may potentially
be at risk [61]. Most clinicians also support the view that
individuals have a right not to know and that this right is
a correlate of the right to know, in regard to both policy
making and genetic counseling. They thus believe that they
must continue to respect patients autonomy and
confidentiality, in order to establish and maintain the trust
between clinicians and patients that is key to the therapeutic
relationship [8, 10, 6972]. Some authors, however, have
argued that the right not to know (or not to share) genetic
information is equivalent to an irresponsible right to
ignorance that may lead to imprudent decisions and cause
harm to oneself and others [61, 71]. Others have appealed to
legal cases, such as the Tarasoff precedent, to advocate for
physicians duty to warn families about genetic risks, stirring an
intense debate about the application of legal precedents to
genetic testing for cancer predisposition [18, 70, 73].
In the USA, the Health Insurance Portability and
Accountability Act (HIPPA) reinforces the ethical and legal
value of confidentiality. In addition, most medical associations
agree that while physicians have a duty to advise patients of the
relevance that genetic information can have to their relatives,
and therefore of the importance to share such information, they
do not have a duty to breach patient confidentiality and warn
their patients relatives directly [62].
Studies have shown that some mutation carriers prefer not to
share their genetic knowledge with family members due to
estranged relationships, unresolved conflicts or uncertainties

symposium article

symposium article
genetic information and its potential relevance to their families,
while refraining from being judgmental about some patients
preferences not to know or not to disclose genetic information
[8, 74]. Over time, physicians may explore with their patients
their motivations related to the nature, meaning and relevance
of a positive genetic test and communication of test results to
family members. Through repeat encounters and through
proper counseling referrals, often patients reach a deeper
understanding of the implications of genetic testing and come
to the decision to share relevant information with other family
members. This, however, may not be the case when patients
and families belong to cultures in which cancer is still
a metaphor of death or of shame and guilt. In such instances,
oncologists should work as part of a team in trying to convey to
their patients a more realistic view of cancer as a potentially
curable or chronic illness, worth preventing or treating as early
as possible [80, 81].

Genetic knowledge can be perceived either as a means to


enhance control of persons lives or as a sign of predestination
written in a mysterious future diary [13, 20]. We therefore
need to create a culture of tolerance, based on appreciation of
all differences, including genetic ones, as an enriching, rather
than impoverishing, element of human society. With respect to
breast cancer, a disease that ranks as second cause of cancer
mortality in women, we must concentrate on identifying new
genetic mutations that predispose patients to breast and other
cancers. We must also become versed in new molecular entities
associated with differential prognosis and response to
antineoplastic therapies in order to refine our predictions and
tailor treatments in a personalized approach to our patients
[85].
The introduction of DTC genetic testing and wholegenome profiling brings new hopes, but they introduce new
ethical consideration, from monitoring the role of economic
interests to the impact on the patientdoctor relationship in
terms of confidentiality and privacy. In the future, oncologists
will be increasingly asked to comment on genetic tests that
they may not have ordered. This will require continuous
update in ones medical education as well as revision, from
time to time, of regulations and legislation. New variables
related to scientific and technological progress need to be
carefully evaluated and openly debated not only within the
scientific and legal communities but also within a more open
forum.
Genetics reminds us of our connectedness to our fellow
human beings. It prompts a reappraisal of the notions of
individual and relational autonomy from which all of us, as
physicians or as actual or potential carriers of genetic mutations
predisposing to cancer susceptibility, may benefit in our moral
understandings. As we enter the era of personalized medicine
based on genomic profiles, we must continue to assure the
safety, reliability and clinical value of all genetic tests offered.
We must also be engaged in protecting patients, their families,
and the public from untoward social and ethical consequences
of genetic testing by adopting and acting on a balanced, nondeterminist, view of genetic information.

i64 | Surbone

disclosures
The author declares no conflict of interest.

references
1. American Cancer Society. Cancer facts and figures [on-line] 2007 http://
www.cancer.org (1 November 2010, date last accessed).
2. Agency for Healthcare Research and Quality. Genetic risk assessment and BRCA
mutation testing for breast and ovarian cancer susceptibility: evidence synthesis
37. Rockville, MD: Agency for Healthcare Research and Quality, 2005.
3. IOM, Institute of Medicine. Assessing Genetic Risks: Implications for Health and
Social Policy. Washington, DC: National Academy Press, 1994.
4. Berliner JL, Fay AM. Risk Assessment and Genetic Counseling for Hereditary
Breast and Ovarian Cancer: Recommendations of the National Society of Genetic
Counselors. Chicago: National Society of Genetic Counselors, Inc., 2007.
5. Patrick-Miller L, Bradbury AR, Terry MB. Controversies in communication of
genetic screening results for cancer: A Report from the American Society of
Preventive Oncologys Screening Special Interest Group. ASPOs 33rd Annual
Meeting, March 2009, Tampa, FL, USA.
6. Rebbeck T, Friebel T, Lynch HT et al. Bilateral prophylactic mastectomy reduces
breast cancer risk in BRCA1 and BRCA2 mutation carriers: the PROSE study
group. J Clin Oncol 2004; 22: 6.
7. Domchek S, Friebel T, Neuhausen SL et al. Mortality reduction after riskreducing bilateral salpingo-oophorectomy in a prospective cohort of BRCA1 and
BRCA2 mutation carriers. Lancet Oncol 2006; 7: 223229.
8. American Society of Clinical Oncology. Statement of the American Society of
Clinical Oncology: genetic testing for cancer susceptibility. J Clin Oncol 1996;
14: 17301736.
9. Burke W, Daly M, Garber J et al. Recommendations for follow-up care of
individuals with an inherited predisposition to cancer. II. BRCA1 and BRCA2.
Cancer Genetics Studies Consortium. JAMA 1997; 277: 9971003.
10. Surbone A. Ethical implications of genetic screening for breast cancer
predisposition. Crit Rev Oncol Hematol 2001; 40: 149157.
11. Chen WY, Garber JE, Higham S et al. BRCA1/2 genetic testing in the community
setting. J Clin Oncol 2002; 20: 4485.
12. Pasacreta JV. Psychosocial issues associated with genetic testing for breast
and ovarian cancer risk: an integrative review. Cancer Invest 2003; 21: 588623.
13. Surbone A. Balance between science and morality. Ann Oncol 2004; 15 (Supp l):
i60i64.
14. Harris M, Winship I, Spriggs M. Controversies and ethical issues in cancergenetics clinics. Lancet Oncol 2005; 6: 301310.
15. Mor P, Oberle K. Ethical issues related to BRCA gene testing in orthodox Jewish
women. Nurs Ethics 2008; 15: 512522.
16. Wang CW, Hui EC. Ethical, legal and social implications of prenatal and
preimplantation genetic testing for cancer susceptibility. Reprod Biomed Online
2009; 19 (Suppl 2): 2333.
17. Offit K, Thom P. Ethicolegal aspects of cancer genetics. Cancer Treat Res 2010;
155: 114.
18. Quinn GP, Vadaparampil ST, Bower B et al. Decisions and ethical issues among
BRCA carriers and the use of preimplantation genetic diagnosis. Minerva Med
2009; 100: 371383.
19. Bradbury AR, Patrick-Miller L, Pawlowski K et al. Should genetic testing for
BRCA1/2 be permitted for minors? Opinions of BRCA mutation carriers and their
adult offspring. Am J Med Genet 2008; 148: 7077.
20. American College of Medical Genetics. ACMG statement on direct-to-consumer
genetic testing [on-line] www.acmg.net (7 April 2008, date last accessed).
21. Matloff E, Caplan A. Direct to confusion: lessons learned from marketing BRCA
testing. Am J Bioeth 2008; 8: 58.
22. Bayeritz K. Whats special about molecular genetic diagnostics? J Med Philos
1998; 23: 247254.
23. Murray TH. Genetic exceptionalism and future diaries: is genetic information
different form other medical information?. In Rothstein MA (ed.), Genetic Secrets:
Protecting Privacy And Confidentiality in the Genetic Era. New Haven, CT: Yale
University Press, 1997.

Volume 22 | Supplement 1 | January 2011

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

conclusion

Annals of Oncology

Annals of Oncology

Volume 22 | Supplement 1 | January 2011

53. Cook-Deegan R, DeRienzo C, Carbone J et al. Impact of gene patents and


licensing practices on access to genetic testing for inherited susceptibility to
cancer: comparing breast and ovarian cancers with colon cancers. Genet Med
2010; 12: S1538.
54. Kuehn BM. Risks and benefits of direct-to-consumer genetic testing remain
unclear. JAMA 2008; 300: 15031505.
55. Leading Edge. Direct-to-consumer genetic tests: flawed and unethical. Lancet
Oncol 2008; 9: 1113.
56. Myers M, Chang M, Jorgensen C et al. Genetic testing for susceptibility to breast
and ovarian cancer: evaluating the impact of a direct-to-consumer marketing
campaign on physicians knowledge and practices. Gen Med 2006; 8:
361370.
57. Samani NJ, Tomaszewski M, Schunkert H. The personal genomethe future of
personalised medicine? Lancet 2010; 375: 14971498.
58. Lucassen A, Parker M. Confidentiality and sharing genetic information with
relatives. Lancet 2010; 375: 15081509.
59. McGuire A, Diaz CM, Hilsenbeck SG. Social networkers attitudes toward directto consumer personal genome testing. Am J Bioeth 2009; 9: 310.
60. Ormond KE, Wheeler M, Hudgins L et al. Challenges in the clinical application of
whole-genome sequencing. Lancet 2010; 375: 17491751.
61. Rhodes R. Genetic links, family ties, and social bonds: rights and responsibilities
in the face of genetic knowledge. J Med Phil 1998; 23: 1030.
62. Offitt K, Groeger E, Turner S. The duty to warn a patients family members
about hereditary disease risks. JAMA 2004; 292: 14691473.
63. Lenoir N. UNESCO, genetics and human rights. Kennedy Inst Ethics J 1997; 7:
3142.
64. Dugan RB, Wiesner GL, Juengst ET et al. Duty to warn at-risk relatives for
genetic disease: genetic counselors clinical experience. Am J Med Genet Part C
(Semin Med Genet) 2003; 119C: 2734.
65. Wertz DC, Fletcher JC. Attitudes of genetic counselors: a multinational survey.
Am J Hum Genet 1988; 42: 592600.
66. Mahowald MB. Genes, Women, Equality. New York: Oxford University Press,
2000.
67. Sherwin S. A relational approach to autonomy in health care. In Sherwin S,
Coordinator, The Feminist Health Care Ethics Research Network, The Politics of
Womens Health: Exploring Agency and Autonomy. Philadelphia: Temple
University Press 1998; 1944.
68. Surbone A. Telling truth to patients with cancer: what is the truth? Lancet Oncol
2006; 7: 944950.
69. Zinberg RE. Genetic testing: is there a right not to know? MCN Am J Matern
Child Nurs 2006; 31: 144.
70. Sulmasy DP. Duty to warn about hereditary disease risk. JAMA 2005; 293:
676.
71. McDougall R. Rethinking the right not to know. Monash Bioeth Rev 2004; 23:
2236.
72. Lehman LS, Weeks JC, Klar N et al. Disclosure of familial genetic information:
perceptions of the duty to inform. Am J Med 2000; 109: 705711.
73. Sulmasy DP. On warning families about genetic risk: the ghost of Tarasoff. Am J
Med 2000; 109: 738739.
74. DAgincourt-Canning L. Experiences of genetic risk: disclosure and the
gendering of responsibility. Bioethics 2001; 15: 231247.
75. Hopwood P. Psychosocial aspects of risk communication and mutation testing in
familial breast-ovarian cancer. Curr Opin Oncol 2005; 17: 340344.
76. Nycum G, Avard D, Knoppers BM. Factors influencing intrafamilial
communication of hereditary breast and ovarian cancer genetic information. Eur
J Hum Gen 2009; 17: 872880.
77. DAgincourt-Canning L, Baird P. Genetic testing for hereditary cancers: the
impact of gender on interest, uptake and ethical considerations. Crit Rev Oncol
Hematol 2006; 58: 114123.
78. Gilbar R. Communicating genetic information in the family:
the familial relationship as the forgotten factor. J Med Ethics 2007; 33:
390393.
79. Wertz DC, Fletcher JC, Nippert I, Wolff G, Ayme S. In focus. Has patient
autonomy gone too far? Geneticists views in 36 nations. Am J Bioeth 2002; 2:
W21.

doi:10.1093/annonc/mdq668 | i65

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

24. Natowicz MR, Alper JK, Aper JS. Genetic discrimination and the law. Am J Hum
Genet 1992; 50: 465475.
25. Billings PR, Kohn MA, deCuevas M et al. Discrimination as a consequence of
genetic testing. Am J Hum Genet 1992; 40: 476482.
26. Billings PR. Genetic nondiscrimination. Nat Genet 2005; 37: 559560.
27. Wolf S. Beyond genetic discrimination: toward the broader harm of geneticism.
J Law Med Ethics 1995; 23: 345353.
28. Harmon A. Insurance fears lead many to shun DNA tests. New York Times,
24 February 2008.
29. Matloff ET, Shappell H, Brierly K et al. What would you do? Specialists
perspectives on cancer genetic testing, prophylactic surgery, and insurance
discrimination. J Clin Oncol 2000; 18: 24842492.
30. Feero G, Guttmacher AE, Collins FS. The genome gets personalalmost. JAMA
2008; 299: 13511352.
31. American Society of Clinical Oncology Policy Statement Update.
genetic and genomic testing for cancer susceptibility. J Clin Oncol 2010; 28:
893901.
32. Kass NE, Medley AM, Natowicz MR et al. Access to health insurance:
experiences and attitudes of those with genetic versus non-genetic medical
conditions. Am J Med Genet 2007; 143: 707717.
33. Wertz DC. Genetic discrimination: an overblown fear? Nat Rev Genet 2002; 3:
496.
34. Nowlan W. A scarlet letter or a red herring? Nature 2003; 421: 313.
35. Taylor S, Treloar S, Barlow-Stewart K et al. Investigating genetic discrimination in
Australia: a large-scale survey of clinical genetics clients. Clin Genet 2008; 74:
2030.
36. Nicolas P. Ethical and juridical issues of genetic testing. A review of the
international regulation. Crit Rev Oncol Hematol 2009; 69: 98107.
37. Van Hoyweghen I, Horstman K. European practices of genetic information and
insurance. Lessons for the Genetic Information Nondiscrimination Act. JAMA
2008; 300: 326327.
38. Bill H. R. 493 [on-line] http://thomas.loc.gov (1 November 2010, date last
accessed).
39. Hudson K, Holohan MK, Collins F. Keeping pace with the times the Genetic
Information Nondiscrimination Act of 2008. N Engl J Med 2008; 358:
26612663.
40. Hampton T. Congress passes bill to ban discrimination based on individuals
genetic makeup. JAMA 2008; 299: 2493.
41. Honey K. GINA: making it safe to know whats in your genes. J Clin Invest 2008;
118: 2369.
42. Annas GJ, Roche P, Green RC. GINA, genism and civil rights. Bioethics 2008;
22: iiiv.
43. Surbone A. Legislation on genetic testing and the practice of oncology. Crit Rev
Oncol Haematol 2009; 69: 9597.
44. The Associated Press. Connecticut woman alleges genetic discrimination at
work. 28 April 2010.
45. Godard B, Raeburn S, Pembrey M et al. Genetic information and testing in
insurance and employment: technical, social and ethical issues. Eur J Hum
Genet 2003; 11 (Suppl 2): 51235142.
46. Alper JS, Beckwith J. Distinguishing genetic from non-genetic medical tests:
some implications for anti-discrimination legislation. Sci Eng Ethics 1998; 4:
141150.
47. Van Hoywegehn I, Horstamn K, Schepers R. Genetic risk carriers and life style
risk takers: which risks deserve our legal protection in insurance? Health Care
Anal 2007; 15: 179193.
48. Hall MA, McEwan JE, Barton JV et al. Concerns in a primary care
population about genetic discrimination by researchers. Genet Med 2005; 7:
311316.
49. Hudson KL. Prohibiting genetic discrimination. New Engl J Med 2007; 356:
20212023.
50. Billings PR. Beyond GINA. Nat Med 2008; 14: 806.
51. Hunter DJ, Khoury MJ, Drazen JM. Letting the genome out of the bottlewill we
get our wish? N Engl J Med 2008; 358: 105107.
52. Offit K. Genomic profiles for disease risk: predictive or premature? JAMA 2008;
299: 13531355.

symposium article

symposium article
80. Loud JT, Weissman NE, Peters JA et al. Deliberate deceit of family members:
a challenge to providers of clinical genetics services. J Clin Oncol 2006; 24:
16431646.
81. Baider L, Surbone A. Cancer and the family: the silent words of truth. J Clin
Oncol 2010: 12691272.
82. Hopwood P, van Asperen C, Legius E et al. Cancer genetics service provision:
a comparison of 7 European centres. Commun Genet 2003; 6: 192205.

Annals of Oncology

83. General Medical Council. Doctors told to warn patients of their genetics risks
[on-line] http://www.gmc-uk.org/guidance/news (28 September 2009, date last
accessed).
84. Parker M, Lucassen AM. Genetic information: a joint account? BMJ 2004; 329:
165167.
85. Huang C-C, Bredel M. Use of gene signatures to improve risk estimation in
cancer. JAMA 2008; 299: 16051606.

Downloaded from http://annonc.oxfordjournals.org/ by guest on April 22, 2012

i66 | Surbone

Volume 22 | Supplement 1 | January 2011

Das könnte Ihnen auch gefallen