Thyroid cancer genetics - Future Medicine

0 downloads 0 Views 710KB Size Report
the human book of life … it is humbling for me and awe inspiring to realize that we have caught the first glimpse of our own instruction book.” – Francis Collins ...
Editorial For reprint orders, please contact: [email protected]

Thyroid cancer genetics: how close are we to personalizing clinical management? “The most successful manner to personalize clinical management is a multi- and inter-disciplinary one, whereby all ’omic networks (genomic, metabolomics and others) are integrated with clinical phenotypes (phenomic), including family health history.”

KEYWORDS: Cowden syndrome n personalized medicine n PTEN mutation n thyroid cancer

“We celebrate the revelation of the first draft of the human book of life … it is humbling for me and awe inspiring to realize that we have caught the first glimpse of our own instruction book.” – Francis Collins, NIH (MD, USA) Over two millennia ago, Hippocrates (460–377 BC) emphasized the importance of individu­ alizing medical care, proclaiming, “It is more important to know what sort of person has a dis­ ease than to know what sort of disease a person has” [101] . Even more poignantly, the alleged first Chinese medical text N’ai Ching (ca 2600 BC) linked prevention with superior doctors and reactive treatment of disease to inferior doctors. Today, advances in genome technologies and the ensuing out­pouring of genomic information related to cancer have led to the convergence of the discovery of science and clinical medicine, allowing clinicians to tailor the management for individual patients. Discoveries in the field of endocrine cancer have contributed much to the early successes we are seeing in personal­ ized healthcare and heralded the way forward for person­alizing management, not only for the patient, but also for their families. While thyroid cancer accounts for approxi­ mately 1% of all newly diagnosed cancer cases, it is the most common malignancy of endocrine organs. Its incidence has increased significantly in the USA and in other countries over the last several decades [1] . This progressive increase can­ not be explained simply by improved diagnostic histo­pathology or preclinical detection through neck ultrasounds; other factors such as environ­ mental influences and molecular alterations must be taken into account [2] . Well differentiated non­ medullary thyroid cancer (NMTC) accounts for 95% of thyroid malignancies and 5% of these

will have familial disease. This contrasts with 10–25% of patients with medullary thyroid cancer (MTC) having a familial etiology [3,4] . Molecular studies have led to increased apprecia­ tion of the heterogeneity of thyroid cancers, with hereditary predisposition, somatic mutation and epigenetic modulation all contributing to tumor behavior [5] . For example, the discovery of the RET proto-oncogene as the causative gene for multiple endocrine neoplasia type-2 allowed for gene-based molecular diagnosis, predictive testing and genotype-specific management for affected individuals. This continues to serve as the para­ digm for personalizing management based on germline mutation status and precise genotype [4,6] . Somatic RET mutations are also found in at least 50% of sporadic MTCs. Importantly, this link between MTC and RET has recently led to the US FDA-approval of vandetanib in 2011, an orally bioavailable inhibitor of RET, VEGFR-2 and EGFR for use in advanced MTC, even those arising in the sporadic setting, follow­ ing dramatic improvements in ­progression-free survival over placebo [7] . Germline mutations in familial NMTC syn­ dromes have not been defined as well as those in MTC. Familial NMTC syndromes include a number of disorders, the commonest of which is Cowden syndrome (CS). As data from the followup of CS patients accumulate, we now know that the lifetime risk of thyroid cancer in CS patients with PTEN mutations is approximately 35% [8] . We demonstrated that the presence of a germline PTEN mutation significantly elevated the risk of epithelial thyroid cancer by approximately 70-fold when compared with the general population [9] . We also saw an increased thyroid cancer risk in CS and CS-like patients with germline SDHx altera­ tions and KLLN promoter hyper­methylation, two

10.2217/PME.12.31 © 2012 Future Medicine Ltd

Personalized Medicine (2012) 9(4), 355–358

Joanne Ngeow Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA and Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA

Charis Eng Author for correspondence: Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, OH 44195, USA and Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA and Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA and Thyroid Cancer Center, Endocrinology & Metabolism Institute, Cleveland Clinic, Cleveland, OH 44195, USA and Stanley Shalom Zielony Institute of Nursing Excellence, Cleveland Clinic, Cleveland, OH 44195, USA and Department of Genetics, & CASE Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA Tel.: +1 216 444 3440 Fax: +1 216 636 0655 [email protected]

part of

ISSN 1741-0541

355

Editorial

Ngeow & Eng

novel genes linked with CS [10,11] . Importantly, our study revealed gene-specific differences in clinical presentations between the three genes, which would suggest the personalization of management in a gene-specific manner. CS and CS-like patients with germline PTEN mutations were more likely to be younger – some present­ ing as early as 7 years of age – and tended to be associated with benign thyroid diseases, such as thyroid adenoma and thyroiditis. There was an over-representation of follicular thyroid cancer (FTC) compared with papillary thyroid cancer (PTC) in patients with germline PTEN muta­ tions, these were distinct from those who had germline SDHx or KLLN alterations. Finding a germline PTEN mutation in an individual would mean increased clinical screening for other asso­ ciated cancers, such as breast and endometrial cancers [8] . Knowing that the renal cancers asso­ ciated with germline PTEN mutations are mainly papillary in histology informs the type of clinical surveillance, namely that the standard ultrasound is insufficient; instead, computed tomography or MRI would be the platform of choice [12] . Individuals with germline SDH mutations have an increased risk of breast and thyroid cancers compared with those with PTEN mutations [13] , while individuals with germline KLLN promoter hyper­methylation have increased risks of breast and renal cancers [10] . This implies that surveil­ lance of CS patients will need to be personalized depending on the underlying germline alteration involved.

“Importantly, our study revealed genespecific differences in clinical presentations between the three genes, which would suggest the personalization of management in a gene-specific manner.” Hereditary thyroid cancer, as illustrated above, provides a model for how personalized healthcare can successfully be applied to the individual and his/her family. Nonetheless, the vast majority of thyroid cancers are sporadic. Sporadic NMTC is characterized by two distinct somatic molecular mechanisms: point mutations or chromosomal rearrangements, which are, in turn, associated with specific etiologic factors involved in thyroid carcinogenesis [5] . There are distinct differences in tumor-mutation profiles between different histo­ logical subtypes. Activating somatic mutations in BRAF (in particular the V600E mutation) have been identified in approximately 50% of PTCs and correlate with poor outcome [14,15] . Somatic RAS mutations, particularly those that involve 356

Personalized Medicine (2012) 9(4)

NRAS and HRAS, are found in 10–20% of PTCs (usually in the follicular variant) and in 40–50% of FTCs [16] . Translocations that result in a fusion between PAX8 and the peroxisome proliferatoractivated receptor g gene are found in 30–40% of FTCs [17] . Although these molecular profiles have not as yet significantly affected the diagnostic prac­ tice of surgical pathology, there is much hope and hype that it can be used to significantly improve the sensitivity of cytological fine-needle aspiration diagnoses of thyroid nodules. Postsurgical results of nodules with ambiguous fine-needle aspiration yields show that only 20–30% of these patients have cancer. We and others have developed pos­ sible gene signatures which show promise in being able to accurately predict thyroid cancer from benign thyroid ­nodules [18,19] . Characterization of these genetic events in thy­ roid cancer may have diagnostic and prognostic implications, but most excitingly, it also provides an opportunity to develop therapies that are tar­ geted at these potential molecular drivers. For patients with MTC treated with vandetanib, a similar response rate was seen in patients with hereditary MTC and in RET mutation-positive sporadic MTC [7] . The M918T mutation (a tyro­ sine kinase domain mutation) was the most fre­ quent somatic mutation found in patients with RET mutation-positive sporadic MTC. The response rate in patients with sporadic MTC and M918T mutations who were treated with vande­ tanib was significantly higher (54 vs 32%) than in patients who were negative for mutations or for whom the mutation status was unknown. This places MTC alongside melanoma and gastro­ intestinal stromal tumors, whereby a woeful lack of efficacy with conventional chemotherapy has now been replaced with renewed hope using targeted therapy.

“Characterization of these genetic events in

thyroid cancer may have diagnostic and prognostic implications, but most excitingly, it also provides an opportunity to develop therapies that are targeted at these potential molecular drivers.” The concept of personalized medicine is not new, but what is new is our ability to utilize an individual’s and the tumor’s genetic make-up to determine clinical management with molecular testing. Over the past two decades, we were able to capitalize on the discovery of RET mutations in multiple endocrine neoplasia type-2 related and sporadic MTC into successful strategies for predictive testing of affected individuals and future science group

Thyroid cancer genetics: how close are we to personalizing clinical management?

hence, genotype-specific clinical surveillance and prophylactic surgery [6] , and along the way, revolutionized treatment for MTC. It is hoped that similar progress can be achieved by a deep­ ening understanding of how germline PTEN mutations, and SDHx and KLLN alterations impact on NMTC tumorigenesis. We await with interest the outcomes of ongoing trials focused on downstream targets of PTEN, such as mTOR and PIK3CA inhibitors that are underway in both CS patients (NCT00971789) as well as in sporadic thyroid cancer with somatic PTEN loss (NCT01430572) [102,103] . We now know that germline SDHx variants may affect mitochon­ drial metabolite dysregulation and subsequent tumorigenesis [13] . While the mechanism(s) of disruption of mitochondrial function leading to neoplasia remain unclear, further research may unravel how we can harness drugs targeting ‘energetics’ for prevention or treatment. As we learn more about the heterogeneity of tumor formation, we are learning the increas­ ing importance of crosstalk between different pathways leading to differential organ-specific carcino­genesis. It is certain that further progress in our understanding of thyroid cancer molecu­ lar genetics will lead to the discovery of novel mutations and other genetic and epigenetic alter­ ations that will help clarify the biology of MTC and NMTC and foster the development of new targeted therapeutic approaches. However, we are going to need to increasingly rethink how we integrate the molecular data that are obtained. In the past, researchers have been approach­ ing the problem in ‘germline’ versus ‘somatic’ silos; however, going forward, it is going to be important that we embrace that tumor biology is likely to be dictated by somatic changes in the cancer multi-‘ome’, the microenvironment, as well as germline ’omic make-up. It is the natural

References 1

2

3

4

tendency to focus on one aspect. The most suc­ cessful manner to personalize clinical manage­ ment is a multi- and inter-disciplinary one, whereby all ’omic networks (genomic, metabo­ lomics and others) are integrated with clinical phenotypes (phenomic), including family health history. While many of us know what must be done to reach the nirvana of person­alized health­ care, there are powerful forces beyond our con­ trol that may derail these noble efforts: lack of research funding, the lack of foresight to invest in this type of bridging research between good ’omics data and clinical implementation (and we do not mean clinical drug trials), and extraordi­ nary regulatory issues, all of which have already stifled our research efforts, at least in the USA. “In expanding the field of knowledge we but increase the horizon of ignorance.” – Henry Miller (1891–1980) Financial & competing interests disclosure Some of the primary research described in this editorial is supported, in part, by grants P01CA124570 and R01CA118980 from the National Cancer Institute, MD, USA and the Breast Cancer Research Foundation (all to C Eng). J Ngeow is the National Medical Research Council (Singapore) Fellow and an Ambrose Monell Foundation Cancer Genomic Medicine Clinical Fellow at the Cleveland Clinic Genomic Medicine Institute. C Eng is the Sondra J and Stephen R Hardis Chair of Cancer Genomic Medicine at the Cleveland Clinic and is an American Cancer Society Clinical Research Professor, generously funded, in part, by the Fred Morgan Kirby Foundation. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials ­discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

5

Davies L, Welch HG. Increasing incidence of thyroid cancer in the United States, 1973–2002. JAMA 295(18), 2164–2167 (2006).

Nikiforov YE, Nikiforova MN. Molecular genetics and diagnosis of thyroid cancer. Nat. Rev. Endocrinol. 7(10), 569–580 (2011).

6

Chen AY, Jemal A, Ward EM. Increasing incidence of differentiated thyroid cancer in the United States, 1988–2005. Cancer 115(16), 3801–3807 (2009).

Kloos RT, Eng C, Evans DB et al. Medullary thyroid cancer: management guidelines of the American thyroid association. Thyroid 19(6), 565–612 (2009).

7

Wells SA Jr, Gosnell JE, Gagel RF et al. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J. Clin. Oncol. 28(5), 767–772 (2010).

Richards ML. Familial syndromes associated with thyroid cancer in the era of personalized medicine. Thyroid 20(7), 707–713 (2010). Zbuk KM, Eng C. Cancer phenomics: RET and PTEN as illustrative models. Nat. Rev. Cancer 7(1), 35–45 (2007).

future science group

8

Editorial

Tan MH, Mester JL, Ngeow J, Rybicki LA, Orloff MS, Eng C. Lifetime cancer risks in individuals with germline PTEN mutations. Clin. Cancer Res. 18(2), 400–407 (2012).

www.futuremedicine.com

9

Ngeow J, Mester J, Rybicki LA, Ni Y, Milas M, Eng C. Incidence and clinical characteristics of thyroid cancer in prospective series of individuals with Cowden and Cowden-like syndrome characterized by germline PTEN, SDH, or KLLN alterations. J. Clin. Endocrinol. Metab. 96(12), E2063–E2071 (2011).

10

Bennett KL, Mester J, Eng C. Germline epigenetic regulation of KILLIN in Cowden and Cowden-like syndrome. JAMA 304(24), 2724–2731 (2010).

11

Ni Y, Zbuk KM, Sadler T et al. Germline mutations and variants in the succinate dehydrogenase genes in Cowden and Cowden-like syndromes. Am. J. Hum. Genet. 83(2), 261–268 (2008).

357

Editorial

Ngeow & Eng

12 Mester JL, Zhou M, Prescott N, Eng C.

Papillary renal cell carcinoma is associated with PTEN hamartoma tumor syndrome. Urology 79(5), 1187.e1–e7 (2012).

15

Xing M. Prognostic utility of BRAF mutation in papillary thyroid cancer. Mol. Cell. Endocrinol. 321(1), 86–93 (2010).

16

Nikiforova MN, Lynch RA, Biddinger PW et al. RAS point mutations and PAX8-PPAR g rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J. Clin. Endocrinol. Metab. 88(5), 2318–2326 (2003).

„„ Websites

Kroll TG, Sarraf P, Pecciarini L et al. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma [corrected]. Science 289(5483), 1357–1360 (2000).

102 Sirolimus to treat Cowden syndrome and

Weber F, Shen L, Aldred MA et al. Genetic classification of benign and malignant thyroid follicular neoplasia based on a three-gene combination. J. Clin. Endocrinol. Metab. 90(5), 2512–2521 (2005).

103 Pazopanib and everolimus in PI3KCA

13 Ni Y, He X, Chen J et al. Germline SDHx

variants modify breast and thyroid cancer risks in Cowden and Cowden-like syndrome via FAD/NAD-dependant destabilization of p53. Hum. Mol. Genet. 21(2), 300–310 (2012). 14

Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA. High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC–RAS–BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 63(7), 1454–1457 (2003).

358

17

18

Personalized Medicine (2012) 9(4)

19

Weber F, Eng C. Update on the molecular diagnosis of endocrine tumors: toward -omics-based personalized healthcare? J. Clin. Endocrinol. Metab. 93(4), 1097–1104 (2008).

101 Alternative medicine online. Quotations.

http://library.thinkquest.org/24206/quotes. html other PTEN hamartomatous tumor syndromes. http://clinicaltrials.gov/ct2/show/ NCT00971789?id=00971789&rank=1 mutation positive/PTEN loss patients. http://clinicaltrials.gov/ct2/show/ NCT01430572?id=01430572&rank=1

future science group