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

Heterogeneity in primary tumors and corresponding metastases: could it provide us with any hints to personalize cancer therapy?

    Ketao Jin

    Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, China

    Department of Surgery, Affiliated Zhuji Hospital, Wenzhou Medical College, Zhuji, Zhejiang, China

    ,
    Kuifeng He

    Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, China

    ,
    Fei Teng

    Faculty of Applied Science, Division of Engineering Science, University of Toronto, Toronto, ON, Canada

    ,
    Na Han

    Sir Run Run Shaw Institute of Clinical Medicine, Zhejiang University, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, China

    ,
    Guangliang Li

    Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, China

    ,
    Zhenzhen Xu

    Sir Run Run Shaw Institute of Clinical Medicine, Zhejiang University, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, China

    &
    Published Online:https://doi.org/10.2217/pme.10.81

    Interpatient variability in response to anticancer drugs is associated with different clinical outcomes, which is partially owing to the individual differences among patients. Many investigators have hoped that tumor heterogeneity would help to reveal the underlying mechanism of interpatient variability in response to anticancer therapy. Numerous studies have demonstrated the presence of intratumor heterogeneity and the heterogeneity in primary tumors and corresponding metastases in a wide range of tumors at different levels and have indicated that the heterogeneity might make sense as a potential determinant of anticancer therapy response. This article discusses tumor heterogeneity, focusing on the heterogeneity in primary tumors and corresponding metastases as well as the effect on anticancer therapy response. Furthermore, an idea of tumor-site-based personalized cancer therapy for patients with metastatic malignancies was hypothesized, and a strategy using a patient-derived tumor tissue xenograft model to realize this idea is also proposed in this article.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Vogel CL, Cobleigh MA, Tripathy D et al.: Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol.20,719–726 (2002).
    • Bell DW, Lynch TJ, Haserlat SM et al.: Epidermal growth factor receptor mutations and gene amplification in non-small-cell lung cancer: molecular analysis of the IDEAL/INTACT gefitinib trials. J. Clin. Oncol.23,8081–8092 (2005).
    • Eberhard DA, Johnson BE, Amler LC et al.: Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J. Clin. Oncol.23,5900–5909 (2005).
    • Bokemeyer C, Bondarenko I, Hartmann JT et al.: KRAS status and efficacy of first-line treatment of patients with metastatic colorectal cancer (mCRC) with FOLFOX with or without cetuximab: the OPUS experience. J. Clin. Oncol.26 (2008) (Abstract 4000).
    • De Roock W, Piessevaux H, De Schutter J et al.: KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. Ann. Oncol.19,508–515 (2008).
    • Arnerlöv C, Emdin SO, Cajander S et al.: Intratumoral variations in DNA ploidy and S-phase fraction in human breast cancer. Anal. Cell Pathol.23,21–28 (2001).
    • Glöckner S, Buurman H, Kleeberger W et al.: Marked intratumoral heterogeneity of c-myc and cyclinD1 but not of c-erbB2 amplification in breast cancer. Lab. Invest.82,1419–1426 (2002).
    • Benetkiewicz M, Piotrowski A, Díaz De Ståhl T et al.: Chromosome 22 array-CGH profiling of breast cancer delimited minimal common regions of genomic imbalances and revealed frequent intra-tumoral genetic heterogeneity. Int. J. Oncol.29,935–945 (2006).
    • Hanna W, Nofech-Mozes S, Kahn HJ: Intratumoral heterogeneity of HER2/neu in breast cancer – a rare event. Breast J.13,122–129 (2007).
    • 10  Brunelli M, Manfrin E, Martignoni G et al.: Genotypic intratumoral heterogeneity in breast carcinoma with HER2/neu amplification: evaluation according to ASCO/CAP criteria. Am. J. Clin. Pathol.131,678–682 (2009).
    • 11  Barry WT, Kernagis DN, Dressman HK et al.: Intratumor heterogeneity and precision of microarray-based predictors of breast cancer biology and clinical outcome. J. Clin. Oncol.28,2198–2206 (2010).
    • 12  Chen C, Peng J, Xia H et al.: Quantum-dot-based immunofluorescent imaging of HER2 and ER provides new insights into breast cancer heterogeneity. Nanotechnology21,095101 (2010).
    • 13  Cottu PH, Asselah J, Lae M et al.: Intratumoral heterogeneity of HER2/neu expression and its consequences for the management of advanced breast cancer. Ann. Oncol.19,595–597 (2008).
    • 14  Lyng MB, Laenkholm AV, Pallisgaard N et al.: Intratumor genetic heterogeneity of breast carcinomas as determined by fine needle aspiration and TaqMan low density array. Cell Oncol.29,361–372 (2007).
    • 15  Sayagués JM, Abad Mdel M, Melchor HB et al.: Intratumoural cytogenetic heterogeneity of sporadic colorectal carcinomas suggests several pathways to liver metastasis. J. Pathol.221,308–319 (2010).
    • 16  Losi L, Baisse B, Bouzourene H et al.: Evolution of intratumoral genetic heterogeneity during colorectal cancer progression. Carcinogenesis26,916–922 (2005).
    • 17  Shen L, Toyota M, Kondo Y et al.: Integrated genetic and epigenetic analysis identifies three different subclasses of colon cancer. Proc. Natl Acad. Sci. USA104,18654–18659 (2007).
    • 18  Bartos JD, Stoler DL, Matsui S et al.: Genomic heterogeneity and instability in colorectal cancer: spectral karyotyping, glutathione transferase-Ml and ras. Mutat. Res.568,283–292 (2004).
    • 19  Half E, Broaddus R, Danenberg KD et al.: HER-2 receptor expression, localization, and activation in colorectal cancer cell lines and human tumors. Int. J. Cancer108,540–548 (2004).
    • 20  Garcia JM, Rodriguez R, Silva J et al.: Intratumoral heterogeneity in microsatellite alterations in BRCA1 and PTEN regions in sporadic colorectal cancer. Ann. Surg. Oncol.10,876–881 (2003).
    • 21  Beau-Faller M, Weber JC, Schneider A et al.: Genetic heterogeneity in lung and colorectal carcinoma as revealed by microsatellite analysis in plasma or tumor tissue DNA. Cancer97,2308–2317 (2003).
    • 22  Whitehall VL, Wynter CV, Walsh MD et al.: Morphological and molecular heterogeneity within nonmicrosatellite instability-high colorectal cancer. Cancer Res.62,6011–6014 (2002).
    • 23  Lassmann S, Bauer M, Soong R et al.: Quantification of CK20 gene and protein expression in colorectal cancer by RT-PCR and immunohistochemistry reveals inter- and intratumour heterogeneity. J. Pathol.198,198–206 (2002).
    • 24  Maekawa M, Sugano K, Ushiama M et al.: Heterogeneity of DNA methylation status analyzed by bisulfite-PCR-SSCP and correlation with clinico-pathological characteristics in colorectal cancer. Clin. Chem. Lab. Med.39,121–128 (2001).
    • 25  Kuwai T, Nakamura T, Kim SJ et al.: Intratumoral heterogeneity for expression of tyrosine kinase growth factor receptors in human colon cancer surgical specimens and orthotopic tumors. Am. J. Pathol.172,358–366 (2008).
    • 26  Blackhall FH, Pintilie M, Wigle DA et al.: Stability and heterogeneity of expression profiles in lung cancer specimens harvested following surgical resection. Neoplasia6,761–767 (2004).
    • 27  Gomez de la Cámara A, López-Encuentra A, Ferrando P et al.: Heterogeneity of prognostic profiles in non-small cell lung cancer: too many variables but a few relevant. Eur. J. Epidemiol.20,907–914 (2005).
    • 28  Perner S, Demichelis F, Beroukhim R et al.: TMPRSS2:ERG fusion-associated deletions provide insight into the heterogeneity of prostate cancer. Cancer Res.66,8337–8341 (2006).
    • 29  Krause FS, Feil G, Bichler KH et al.: Heterogeneity in prostate cancer: prostate specific antigen (PSA) and DNA cytophotometry. Anticancer Res.25,1783–1785 (2005).
    • 30  Chau CH, Figg WD: Molecular and phenotypic heterogeneity of metastatic prostate cancer. Cancer Biol. Ther.4,166–167 (2005).
    • 31  Walsh PC: Heterogeneity of genetic alterations in prostate cancer: evidence of the complex nature of the disease. J. Urol.168,1635–1636 (2002).
    • 32  Nwosu V, Carpten J, Trent JM et al.: Heterogeneity of genetic alterations in prostate cancer: evidence of the complex nature of the disease. Hum. Mol. Genet.10,2313–2318 (2001).
    • 33  Cooke SL, Ng CK, Melnyk N et al.: Genomic analysis of genetic heterogeneity and evolution in high-grade serous ovarian carcinoma. Oncogene29,4905–4913 (2010).
    • 34  Gershenson DM: The heterogeneity of epithelial ovarian cancer: getting it right. Cancer116,1400–1402 (2010).
    • 35  Bast RC Jr, Hennessy B, Mills GB: The biology of ovarian cancer: new opportunities for translation. Nat. Rev. Cancer9,415–428 (2009).
    • 36  McAlpine JN, Eisenkop SM, Spirtos NM: Tumor heterogeneity in ovarian cancer as demonstrated by in vitro chemoresistance assays. Gynecol. Oncol.110,360–364 (2008).
    • 37  Woloszynska-Read A, Mhawech-Fauceglia P, Yu J et al.: Intertumor and intratumor NY-ESO-1 expression heterogeneity is associated with promoter-specific and global DNA methylation status in ovarian cancer. Clin. Cancer Res.14,3283–3290 (2008).
    • 38  Jochumsen KM, Tan Q, Hølund B et al.: Gene expression in epithelial ovarian cancer: a study of intratumor heterogeneity. Int. J. Gynecol. Cancer17,979–985 (2007).
    • 39  Khalique L, Ayhan A, Weale ME et al.: Genetic intra-tumour heterogeneity in epithelial ovarian cancer and its implications for molecular diagnosis of tumours. J. Pathol.211,286–295 (2007).
    • 40  Pieretti M, Hopenhayn-Rich C, Khattar NH et al.: Heterogeneity of ovarian cancer: relationships among histological group, stage of disease, tumor markers, patient characteristics, and survival. Cancer Invest.20,11–23 (2002).
    • 41  Manahan KJ, Taylor DD, Gerçel-Taylor C: Clonal heterogeneity of p53 mutations in ovarian cancer. Int. J. Oncol.19,387–394 (2001).
    • 42  Nakamura T, Kuwai T, Kitadai Y et al.: Zonal heterogeneity for gene expression in human pancreatic carcinoma. Cancer Res.67,7597–7604 (2007).
    • 43  Harada T, Okita K, Shiraishi K et al.: Interglandular cytogenetic heterogeneity detected by comparative genomic hybridization in pancreatic cancer. Cancer Res.62,835–839 (2002).
    • 44  Osterheld MC, Caron L, Demierre M et al.: DNA-ploidy in advanced gastric carcinoma is less heterogeneous than in early gastric cancer. Cell Oncol.26,21–29 (2004).
    • 45  Awata H, Hirose K, Yamaguchi A: Heterogeneity of apoptosis and proliferative activity within superficial spreading type early gastric cancer. Oncol. Rep.9,153–158 (2002).
    • 46  Saito A, Shimoda T, Nakanishi Y et al.: Histologic heterogeneity and mucin phenotypic expression in early gastric cancer. Pathol. Int.51,165–171 (2001).
    • 47  Kobayashi K, Kitayama Y, Igarashi H et al.: Intratumor heterogeneity of centromere numerical abnormality in multiple primary gastric cancers: application of fluorescence in situ hybridization with intermittent microwave irradiation on paraffin-embedded tissue. Jpn. J. Cancer Res.91,1134–1341 (2000).
    • 48  Donahue MJ, Blakeley JO, Zhou J et al.: Evaluation of human brain tumor heterogeneity using multiple T1-based MRI signal weighting approaches. Magn. Reson. Med.59,336–344 (2008).
    • 49  Ren ZP, Olofsson T, Qu M et al.: Molecular genetic analysis of p53 intratumoral heterogeneity in human astrocytic brain tumors. J. Neuropathol. Exp. Neurol.66,944–954 (2007).
    • 50  Wyss MT, Hofer S, Hefti M et al.: Spatial heterogeneity of low-grade gliomas at the capillary level: a PET study on tumor blood flow and amino acid uptake. J. Nucl. Med.48,1047–1052 (2007).
    • 51  Pauliah M, Saxena V, Haris M et al.: Improved T(1)-weighted dynamic contrast-enhanced MRI to probe microvascularity and heterogeneity of human glioma. Magn. Reson. Imaging25,1292–1299 (2007).
    • 52  Andersson U, Malmer B, Bergenheim AT et al.: Heterogeneity in the expression of markers for drug resistance in brain tumors. Clin. Neuropathol.23,21–27 (2004).
    • 53  Walker C, du Plessis DG, Joyce KA et al.: Phenotype versus genotype in gliomas displaying inter- or intratumoral histological heterogeneity. Clin. Cancer Res.9,4841–4851 (2003).
    • 54  Staller P: Genetic heterogeneity and chromatin modifiers in renal clear cell carcinoma. Future Oncol.6,897–900 (2010).▪ Demonstrated the prevalence of discrepant results of HER2 status between the primary breast cancer and the distant metastases.
    • 55  Gong Y, Booser DJ, Sneige N: Comparison of HER-2 status determined by fluorescence in situ hybridization in primary and metastatic breast carcinoma. Cancer103,1763–1769 (2005).
    • 56  Gancberg D, Di Leo A, Cardoso F et al.: Comparison of HER-2 status between primary breast cancer and corresponding distant metastatic sites. Ann. Oncol.13,1036–1043 (2002).
    • 57  Regitnig P, Schippinger W, Lindbauer M et al.: Change of HER-2/neu status in a subset of distant metastases from breast carcinomas. J. Pathol.203,918–926 (2004).
    • 58  Bozzetti C, Personeni N, Nizzoli R et al.: HER-2/neu amplification by fluorescence in situ hybridization in cytologic samples from distant metastatic sites of breast carcinoma. Cancer99,310–315 (2003).
    • 59  Tanner M, Järvinen P, Isola J: Amplification of HER-2/neu and topoisomerase IIα in primary and metastatic breast cancer. Cancer Res.61,5345–5348 (2001).
    • 60  Tapia C, Savic S, Wagner U et al.: HER2 gene status in primary breast cancers and matched distant metastases. Breast Cancer Res.9,R31 (2007).▪ Demonstrated the poor concordance between immunohistochemistry findings of pAkt and p4E-BP1 expression in primary breast cancer and metastases.
    • 61  Akcakanat A, Sahin A, Shaye AN et al.: Comparison of Akt/mTOR signaling in primary breast tumors and matched distant metastases. Cancer112,2352–2358 (2008).▪ Comprehensive comparison of biomarker expression between patients’ primary breast carcinoma and their metastatic breast carcinomas.
    • 62  Wu JM, Fackler MJ, Halushka MK et al.: Heterogeneity of breast cancer metastases: comparison of therapeutic target expression and promoter methylation between primary tumors and their multifocal metastases. Clin. Cancer Res.14,1938–1946 (2008).▪ Demonstrated the heterogeneity of EGFR gene status and its downstream signaling proteins between primary colorectal cancer and its corresponding metastases.
    • 63  Baldus SE, Schaefer KL, Engers R et al.: Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin. Cancer Res.16,790–799 (2010).
    • 64  Molinari F, Martin V, Saletti P et al.: Differing deregulation of EGFR and downstream proteins in primary colorectal cancer and related metastatic sites may be clinically relevant. Br. J. Cancer100,1087–1094 (2009).
    • 65  Scartozzi M, Bearzi I, Berardi R et al.: Epidermal growth factor receptor (EGFR) status in primary colorectal tumors does not correlate with EGFR expression in related metastatic sites: implications for treatment with EGFR-targeted monoclonal antibodies. J. Clin. Oncol.22,4772–4778 (2004).
    • 66  Scartozzi M, Bearzi I, Berardi R et al.: Epidermal growth factor receptor (EGFR) downstream signalling pathway in primary colorectal tumours and related metastatic sites: optimising EGFR-targeted treatment options. Br. J. Cancer97,92–97 (2007).▪ Demonstrated genetic heterogeneity between primary non-small-cell lung cancer and lymph node metastases.
    • 67  Sasatomi E, Finkelstein SD, Woods JD et al.: Comparison of accumulated allele loss between primary tumor and lymph node metastasis in stage II non-small cell lung carcinoma: implications for the timing of lymph node metastasis and prognostic value. Cancer Res.62,2681–2689 (2002).
    • 68  Park S, Holmes-Tisch AJ, Cho EY et al.: Discordance of molecular biomarkers associated with epidermal growth factor receptor pathway between primary tumors and lymph node metastasis in non-small cell lung cancer. J. Thorac. Oncol.4,809–815 (2009).
    • 69  Monaco SE, Nikiforova MN, Cieply K et al.: A comparison of EGFR and KRAS status in primary lung carcinoma and matched metastases. Hum. Pathol.41,94–102 (2010).▪▪ Retrospective study which investigated the role of PTEN loss, AKT phosphorylation and KRAS mutations in primary colorectal tumors and their corresponding metastases on the activity of cetuximab plus irinotecan.
    • 70  Loupakis F, Pollina L, Stasi I et al.: PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. J. Clin. Oncol.27,2622–2629 (2009).
    • 71  Zeimet AG, Marth C: Why did p53 gene therapy fail in ovarian cancer? Lancet Oncol.4,415–422 (2003).
    • 72  Germano IM, Binello E: Gene therapy as an adjuvant treatment for malignant gliomas: from bench to bedside. J. Neurooncol.93,79–87 (2009).
    • 73  Jin K, He K, Li G et al.: Personalized cancer therapy using a patient-derived tumor tissue xenograft model: a translational field worthy of exploring further? Pers. Med.7,597–606 (2010).
    • 74  Dong X, Guan J, English JC et al.: Patient-derived first generation xenografts of non-small cell lung cancers: promising tools for predicting drug responses for personalized chemotherapy. Clin. Cancer Res.16,1442–1451 (2010).