We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Tumor heterogeneity and personalized cancer medicine: are we being outnumbered?

    Geert A Cirkel

    Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands

    ,
    Christa G Gadellaa-van Hooijdonk

    Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands

    ,
    Marco J Koudijs

    Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands

    ,
    Stefan M Willems

    Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands

    &
    Emile E Voest

    * Author for correspondence:

    E-mail Address: e.voest@nki.nl

    Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands

    Published Online:https://doi.org/10.2217/fon.13.214

    ABSTRACT: 

    Tumor heterogeneity is regarded as a major obstacle to successful personalized cancer medicine. The lack of reliable response assays reflective of in vivo tumor heterogeneity and associated resistance mechanisms hampers identification of reliable biomarkers. By contrast, oncogene addiction and paracrine signaling enable systemic responses despite tumor heterogeneity. This strengthens researchers in their efforts towards personalized cancer medicine. Given the fact that tumor heterogeneity is an integral part of cancer evolution, diagnostic tools need to be developed in order to better understand the dynamics within a tumor. Ultra-deep sequencing may reveal future resistant clones within a (liquid) tumor biopsy. On-treatment biopsies may provide insight into intrinsic or acquired drug resistance. Subsequently, upfront combinatorial treatment or sequential therapy strategies may forestall drug resistance and improve patient outcome. Finally, innovative response assays, such as organoid cultures or patient-derived tumor xenografts, provide an extra dimension to correlate molecular profiles with drug efficacy and control cancer growth.

    Papers of special note have been highlighted as:

    • of interest

    •• of considerable interest

    References

    • 1 Schuster-Bockler B, Lehner B. Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature 488(7412), 504–507 (2012).
    • 2 Aguilera A, Gomez-Gonzalez B. Genome instability: a mechanistic view of its causes and consequences. Nat. Rev. Genet. 9(3), 204–217 (2008).
    • 3 Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455(7216), 1061–1068 (2008).
    • 4 Keats JJ, Chesi M, Egan JB et al. Clonal competition with alternating dominance in multiple myeloma. Blood 120(5), 1067–1076 (2012).
    • 5 Ding L, Ley TJ, Larson DE et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481(7382), 506–510 (2012).
    • 6 Hunter C, Smith R, Cahill DP et al. A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy. Cancer Res. 66(8), 3987–3991 (2006).
    • 7 Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 144(5), 646–674 (2011).
    • 8 Yachida S, Jones S, Bozic I et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467(7319), 1114–1117 (2010).
    • 9 Vermaat JS, Nijman IJ, Koudijs MJ et al. Primary colorectal cancers and their subsequent hepatic metastases are genetically different: implications for selection of patients for targeted treatment. Clin. Cancer Res. 18(3), 688–699 (2012).
    • 10 Shah SP, Roth A, Goya R et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 486(7403), 395–399 (2012).
    • 11 Gerlinger M, Rowan AJ, Horswell S et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366(10), 883–892 (2012).
    • 12 Parsons DW, Jones S, Zhang X et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321(5897), 1807–1812 (2008).
    • 13 Jones S, Zhang X, Parsons DW et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321(5897), 1801–1806 (2008).
    • 14 Kloosterman W, Hoogstraat M, Paling O et al. Chromothripsis is a common mechanism driving genomic rearrangements in primary and metastatic colorectal cancer. Genome Biol. 12(10), R103 (2011).
    • 15 Seligson DB, Horvath S, McBrian MA et al. Global levels of histone modifications predict prognosis in different cancers. Am. J. Pathol. 174(5), 1619–1628 (2009).
    • 16 Fraga MF, Ballestar E, Villar-Garea A et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat. Genet. 37(4), 391–400 (2005).
    • 17 Magee JA, Piskounova E, Morrison SJ. Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer Cell 21(3), 283–296 (2012).
    • 18 Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat. Rev. Genet. 7(1), 21–33 (2006).
    • 19 Sharma SV, Lee DY, Li B et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141(1), 69–80 (2010). •• Reports on an acquired, reversible, chromatin-mediated, resistance mechanism in cancer cell subpopulations after exposure to various anticancer drugs. Propagation of resistant cells in a drug-free medium resensitized these cells, providing a rationale to further explore intermitting or alternating dosing regimens.
    • 20 Gupta PB, Fillmore CM, Jiang G et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146(4), 633–644 (2011).
    • 21 Oltedal S, Aasprong OG, Moller JH et al. Heterogeneous distribution of K-ras mutations in primary colon carcinomas: implications for EGFR-directed therapy. Int. J. Colorectal Dis. 26(10), 1271–1277 (2011).
    • 22 Watanabe T, Kobunai T, Yamamoto Y et al. Heterogeneity of KRAS status may explain the subset of discordant KRAS status between primary and metastatic colorectal cancer. Dis. Colon Rectum 54(9), 1170–1178 (2011).
    • 23 Baas JM, Krens LL, Guchelaar HJ, Morreau H, Gelderblom H. Concordance of predictive markers for EGFR inhibitors in primary tumors and metastases in colorectal cancer: a review. Oncologist 16(9), 1239–1249 (2011).
    • 24 Poste G. Bring on the biomarkers. Nature 469(7329), 156–157 (2011).
    • 25 Flaherty KT, Puzanov I, Kim KB et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N. Engl. J. Med. 363(9), 809–819 (2010).
    • 26 Kwak EL, Bang YJ, Camidge DR et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N. Engl. J. Med. 363(18), 1693–1703 (2010).
    • 27 Weinstein IB: Addiction to oncogenes – the Achilles heal of cancer. Science 297(5578), 63–64 (2002). • Addresses the phenomenon of oncogene addiction as a physiological dependency of cancer cells on the continued activity of specifically activated or overexpressed oncogenes for maintenance of their malignant phenotype.
    • 28 Yancovitz M, Litterman A, Yoon J et al. Intra- and inter-tumor heterogeneity of BRAF(V600E))mutations in primary and metastatic melanoma. PLoS ONE 7(1), e29336 (2012).
    • 29 Wilmott JS, Menzies AM, Haydu LE et al. BRAFV600E protein expression and outcome from BRAF inhibitor treatment in BRAFV600E metastatic melanoma. Br. J. Cancer 108(4), 924–931 (2013).
    • 30 Long GV, Wilmott JS, Capper D et al. Immunohistochemistry is highly sensitive and specific for the detection of V600E BRAF mutation in melanoma. Am. J. Surg. Pathol. 37, 61–65 (2013).
    • 31 Krueger DA, Care MM, Holland K et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N. Engl. J. Med. 363(19), 1801–1811 (2010).
    • 32 Fong PC, Boss DS, Yap TA et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361(2), 123–134 (2009).
    • 33 Hammond ME, Hayes DF, Dowsett M et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in Breast Cancer. J. Clin. Oncol. 28(16), 2784–2795 (2010).
    • 34 Barnes DM, Harris WH, Smith P, Millis RR, Rubens RD. Immunohistochemical determination of oestrogen receptor: comparison of different methods of assessment of staining and correlation with clinical outcome of breast cancer patients. Br. J. Cancer 74(9), 1445–1451 (1996).
    • 35 Fillmore CM, Gupta PB, Rudnick JA et al. Estrogen expands breast cancer stem-like cells through paracrine FGF/Tbx3 signaling. Proc. Natl Acad. Sci. USA 107(50), 21737–21742 (2010). •• Reports on an estrogen-induced paracrine FGF/FGF receptor/Tbx3 signaling pathway, which expands the functional cancer stem cell pool in breast cancer. This finding provides a molecular rationale on how a subgroup of estrogen receptor-positive cells targeted with antihormone therapy may influence tumor behavior as a whole.
    • 36 Inda MM, Bonavia R, Mukasa A et al. Tumor heterogeneity is an active process maintained by a mutant EGFR-induced cytokine circuit in glioblastoma. Genes Dev. 24(16), 1731–1745 (2010).
    • 37 Einhorn LH. Curing metastatic testicular cancer. Proc. Natl Acad. Sci. USA 99(7), 4592–4595 (2002).
    • 38 McIntyre A, Summersgill B, Lu YJ et al. Genomic copy number and expression patterns in testicular germ cell tumours. Br. J. Cancer 97(12), 1707–1712 (2007).
    • 39 Brait M, Maldonado L, Begum S et al. DNA methylation profiles delineate epigenetic heterogeneity in seminoma and non-seminoma. Br. J. Cancer 106(2), 414–423 (2012).
    • 40 Shenghui H, Nakada D, Morrison SJ. Mechanisms of stem cell self-renewal. Annu. Rev. Cell Dev. Biol. 25(1), 377–406 (2009).
    • 41 Tomasetti C, Vogelstein B, Parmigiani G. Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation. Proc. Natl Acad. Sci. USA 110(6), 1999–2004 (2013).
    • 42 Taniguchi K, Okami J, Kodama K, Higashiyama M, Kato K. Intratumor heterogeneity of epidermal growth factor receptor mutations in lung cancer and its correlation to the response to gefitinib. Cancer Sci. 99(5), 929–935 (2008).
    • 43 Lee AJ, Endesfelder D, Rowan AJ et al. Chromosomal instability confers intrinsic multidrug resistance. Cancer Res. 71(5), 1858–1870 (2011).
    • 44 Kennecke H, Yerushalmi R, Woods R et al. Metastatic behavior of breast cancer subtypes. J. Clin. Oncol. 28(20), 3271–3277 (2010).
    • 45 Turner NC, Reis-Filho JS. Genetic heterogeneity and cancer drug resistance. Lancet Oncol. 13(4), e178–e185 (2012).
    • 46 Sun Y, Nelson PS. Molecular pathways: involving microenvironment damage responses in cancer therapy resistance. Clin. Cancer Res. 18(15), 4019–4025 (2012).
    • 47 Komarova NL, Wodarz D. Drug resistance in cancer: principles of emergence and prevention. Proc. Natl Acad. Sci. USA102(27), 9714–9719 (2005).
    • 48 Das Thakur M, Salangsang F, Landman AS et al. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature 494(7436), 251–255 (2013). • Reports on vemurafenib-resistant patient-derived primary melanoma xenograft models being drug dependent for their continued proliferation. This finding provides a rationale for intermittent vemurafenib dosing, which indeed prolonged progression-free survival in tumor-bearing mice.
    • 49 Prahallad A, Sun C, Huang S et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483(7387), 100–103 (2012).
    • 50 Gotink KJ, Broxterman HJ, Labots M et al. Lysosomal sequestration of sunitinib: a novel mechanism of drug resistance. Clin. Cancer Res. 17(23), 7337–7346 (2011).
    • 51 Piscazzi A, Costantino E, Maddalena F et al. Activation of the RAS/RAF/ERK signaling pathway contributes to resistance to sunitinib in thyroid carcinoma cell lines. J. Clin. Endocrinol. Metab. 97(6), E898–E906 (2012).
    • 52 Hazlehurst LA, Landowski TH, Dalton WS. Role of the tumor microenvironment in mediating de novo resistance to drugs and physiological mediators of cell death. Oncogene 22(47), 7396–7402 (2003).
    • 53 Corcoran RB, Ebi H, Turke AB et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition. Cancer Discov. 2(3), 227–235 (2012).
    • 54 Glasspool RM, Teodoridis JM, Brown R. Epigenetics as a mechanism driving polygenic clinical drug resistance. Br. J. Cancer 94(8), 1087–1092 (2006).
    • 55 Cervera E, Candelaria M, Lopez-Navarro O et al. Epigenetic therapy with hydralazine and magnesium valproate reverses imatinib resistance in patients with chronic myeloid leukemia. Clin. Lymphoma Myeloma Leuk. 12(3), 207–212 (2012).
    • 56 Elias MH, Baba AA, Husin A et al. HOXA4 gene promoter hypermethylation as an epigenetic mechanism mediating resistance to imatinib mesylate in chronic myeloid leukemia patients. Biomed. Res. Int. 2013, 129715 (2013).
    • 57 Yu W, Jin C, Lou X et al. Global analysis of DNA methylation by Methyl-Capture sequencing reveals epigenetic control of cisplatin resistance in ovarian cancer cell. PLoS ONE 6(12), e29450 (2011).
    • 58 Magnani L, Stoeck A, Zhang X et al. Genome-wide reprogramming of the chromatin landscape underlies endocrine therapy resistance in breast cancer. Proc. Natl Acad. Sci. USA 110(16), E1490–E1499 (2013).
    • 59 Diaz LA Jr, Williams RT, Wu J et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486(7404), 537–540 (2012).
    • 60 Forshew T, Murtaza M, Parkinson C et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 4(136), 136ra68 (2012). • Reports on the feasibility of identifying cancer mutations present in circulating DNA at allele frequencies as low as 2%, with high sensitivity and specificity by using a specific method for tagged-amplicon deep sequencing.
    • 61 Gatenby RA, Silva AS, Gillies RJ, Frieden BR. Adaptive therapy. Cancer Res. 69(11), 4894–4903 (2009).
    • 62 Ellis LM, Fidler IJ. Finding the tumor copycat: therapy fails, patients don’t. Nat. Med. 16(9), 974–975 (2010).
    • 63 Begley CG, Ellis LM. Drug development: raise standards for preclinical cancer research. Nature 483(7391), 531–533 (2012). • Addresses the remarkable low number of preclinical findings being translated and validated in daily practice despite major advances in our understanding of cancer biology. The authors provide a set of recommendations in order to improve the reliability of preclinical cancer studies.
    • 64 Bertotti A, Migliardi G, Galimi F et al. A molecularly annotated platform of patient-derived xenografts (“xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 1(6), 508–523 (2011).
    • 65 Kopetz S, Lemos R, Powis G. The promise of patient-derived xenografts: the best laid plans of mice and men. Clin. Cancer Res. 18(19), 5160–5162 (2012).
    • 66 Hidalgo M, Bruckheimer E, Rajeshkumar NV et al. A pilot clinical study of treatment guided by personalized tumor grafts in patients with advanced cancer. Mol. Cancer Ther. 10(8), 1311–1316 (2011).
    • 67 Sato T, Vries RG, Snippert HJ et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459(7244), 262–265 (2009).
    • 68 Sato T, Stange DE, Ferrante M et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology 141(5), 1762–1772 (2011). • Reports on culture conditions that allow growth of tissue from the human GI tract (including primary human colon cancer cells) into organoid cultures. The organoid culture platform may be used for pharmacologic studies thereby potentially facilitating personalized cancer medicine.
    • 69 Kuratnik A, Giardina C. Intestinal organoids as tissue surrogates for toxicological and pharmacological studies. Biochem. Pharmacol. 85(12), 1721–1726 (2013).
    • 70 Jin K, Teng L, Shen Y, He K, Xu Z, Li G. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review. Clin. Transl. Oncol. 12(7), 473–480 (2010).
    • 71 Aparicio S, Caldas C. The implications of clonal genome evolution for cancer medicine. N. Engl. J. Med. 368(9), 842–851 (2013).
    • 72 Hou HW, Warkiani ME, Khoo BL et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 3, 1259 (2013).
    • 73 Nagrath S, Sequist LV, Maheswaran S et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 450(7173), 1235–1239 (2007).
    • 74 Zong C, Lu S, Chapman AR, Xie XS. Genome-wide detection of single-nucleotide and copy-number variations of a single human cell. Science 338(6114), 1622–1626 (2012).
    • 75 Murtaza M, Dawson SJ, Tsui DWY et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497(7447), 108-112 ( 2013).
    • 76 McBride DJ, Orpana AK, Sotiriou C et al. Use of cancer-specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumors. Genes Chromosomes Cancer 49(11), 1062–1069 (2010).
    • 77 Leary RJ, Kinde I, Diehl F et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci. Transl. Med. 2(20), 20ra14 (2010).
    • 78 Diehl F, Schmidt K, Choti MA et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 14(9), 985–990 (2008).
    • 79 Center for Personalized Cancer Treatment. www.cpct.nl
    • 80 DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of drug development costs. J. Health Econ. 22(2), 151–185 (2003).
    • 81 Lin NU, Winer EP. Chemotherapy agents in human epidermal growth factor receptor 2 positive breast cancer: time to step out of the limelight. J. Clin. Oncol. 29(3), 251–253 (2011).