The surgical intelligent knife distinguishes normal, borderline and malignant gynaecological tissues using rapid evaporative ionisation mass spectrometry (REIMS)

David L Phelps, Júlia Balog, Louise F Gildea, Zsolt Bodai, Adele Savage, Mona A El-Bahrawy, Abigail Vm Speller, Francesca Rosini, Hiromi Kudo, James S McKenzie, Robert Brown, Zoltán Takáts, Sadaf Ghaem-Maghami, David L Phelps, Júlia Balog, Louise F Gildea, Zsolt Bodai, Adele Savage, Mona A El-Bahrawy, Abigail Vm Speller, Francesca Rosini, Hiromi Kudo, James S McKenzie, Robert Brown, Zoltán Takáts, Sadaf Ghaem-Maghami

Abstract

Background: Survival from ovarian cancer (OC) is improved with surgery, but surgery can be complex and tumour identification, especially for borderline ovarian tumours (BOT), is challenging. The Rapid Evaporative Ionisation Mass Spectrometric (REIMS) technique reports tissue histology in real-time by analysing aerosolised tissue during electrosurgical dissection.

Methods: Aerosol produced during diathermy of tissues was sampled with the REIMS interface. Histological diagnosis and mass spectra featuring complex lipid species populated a reference database on which principal component, linear discriminant and leave-one-patient-out cross-validation analyses were performed.

Results: A total of 198 patients provided 335 tissue samples, yielding 3384 spectra. Cross-validated OC classification vs separate normal tissues was high (97·4% sensitivity, 100% specificity). BOT were readily distinguishable from OC (sensitivity 90.5%, specificity 89.7%). Validation with fresh tissue lead to excellent OC detection (100% accuracy). Histological agreement between iKnife and histopathologist was very good (kappa 0.84, P < 0.001, z = 3.3). Five predominantly phosphatidic acid (PA(36:2)) and phosphatidyl-ethanolamine (PE(34:2)) lipid species were identified as being significantly more abundant in OC compared to normal tissue or BOT (P < 0.001, q < 0.001).

Conclusions: The REIMS iKnife distinguishes gynaecological tissues by analysing mass-spectrometry-derived lipidomes from tissue diathermy aerosols. Rapid intra-operative gynaecological tissue diagnosis may improve surgical care when histology is unknown, leading to personalised operations tailored to the individual.

Conflict of interest statement

Z.T. serves as a paid consultant for Waters Corporation. J.B. is employed by Waters Corporation. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1
REIMS set-up ex-vivo and in operating theatres (in-vivo) and frozen sample work-flow. a Electrical current, produced from the generator, is applied to the tissue and the resultant charged particles are extracted through the custom-designed hand-piece and drawn into the REIMS atmospheric inlet and analysed in the Xevo G2-XS mass spectrometer to produce tissue-specific mass spectra, which are then subjected to multivariate statistical analysis using PC-LDA. Within one to two seconds, real-time tissue diagnosis is displayed on a screen for the surgeon to see. b Work-flow for the frozen samples; all samples collected from the frozen tissue bank were processed with the iKnife. After histopathology reporting 22 samples were rejected from the study due to not being gynaecological or epithelial ovarian samples, or the histology was unclear and they failed quality control (QC). The remainder of the samples (n = 150) and resultant spectra were included in subsequent models, univariate and multivariate analyses
Fig. 2
Fig. 2
Multivariate analyses of ovarian cancer and normal tissue types. a 3-component 3D PCA, percentage of variance explained in PCs1–3: 36.0, 17.6, 9.0%. b 25-component 3D PC-LDA. c Box plots of univariate analysis showing the top m/z peaks contributing to class separation (log2 fold change), between ovarian cancer and normal samples combined. Ten most significant and intense REIMS spectra peaks shown with P < 0.001, q < 0.001. Figure based on the following sample numbers: ovarian cancer n = 39, normal ovary n = 15, normal fallopian tube n = 15, normal peritoneum n = 14
Fig. 3
Fig. 3
Multivariate statistical analyses of ovarian cancer, benign and borderline ovarian tumours. a 2-component PCA, b 25-component 2D PC-LDA, c Leave-one-patient-out cross-validation when all three tissue classes are included in the model, d Box plots of univariate analysis showing the top m/z peaks contributing to class separation (log2 fold change), between ovarian cancer and borderline ovarian tumours. Ten most significant and intense REIMS spectra peaks shown with P < 0.001, q < 0.001, eg Binary LOPOCV models showing improved classification when only two classes are included in the model. Number of samples: ovarian cancer n = 39, benign n = 22, borderline tumour n = 21. Only OC samples with at least 50% of viable tumour were included
Fig. 4
Fig. 4
Validation of the frozen tissue model using fresh tissue. a 2D PCA model of frozen ovarian cancer samples (>50% tumour content) and all normal samples combined as ‘Normal Tissue’. b 3D LDA component analysis. c Leave-one-patient-out cross-validation of the binary ‘Ovarian Cancer’ vs ‘Normal Tissue’ frozen model, showing 100% correct classification. d OMB Recognition software example showing reported classification for individual sampling points (burns) and the probability value associated with the reported class. e Bar chart showing the numbers of sampling points in fresh tissue classified with >75% probability using the OMB recognition software, plus the diagnostic accuracy at those sampling points [OC 100% (61/61), normal ovary 100% (64/64), fallopian tube 100% (58/58), peritoneum 94.1% (32/34)]
Fig. 5
Fig. 5
Spatial resolution of metastatic peritoneal deposits and iKnife recognition. a Peritoneum containing metastatic tumour deposits with iKnife burns labelled 1–16 sampling normal peritoneum and tumour nodules. b Matched haematoxylin and eosin histological slide for the tissue sample in panel A with corresponding burn sites numbered. Scale showing millimetres. c Total ion chromatogram obtained during the sampling of the specimen with the iKnife with each burn numbered. Coloured boxes represent data obtained from a nodule (red; burns 1&2) and normal peritoneum (green; burns 9&10). d Representative mass spectra, obtained in negative-ion mode from a nodule (red) and normal peritoneum (green) showing the degree of variability in the MS peaks for different tissue types (OC vs normal). e Surgeon’s histological impression, iKnife’s impression (percentages in parenthesis represent probability of correct classification) and the histopathologist’s final diagnosis (percentages in parenthesis represent tumour cell content)

References

    1. Agarwal R, Kaye SB. Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nat. Rev. Cancer. 2003;3:502–516. doi: 10.1038/nrc1123.
    1. ACS (2016) Survival Rates for Ovarian Cancer, by Stage, Vol. 2017,
    1. Chang SJ, Hodeib M, Chang J, Bristow RE. Survival impact of complete cytoreduction to no gross residual disease for advanced-stage ovarian cancer: a meta-analysis. Gynecol. Oncol. 2013;130:493–498. doi: 10.1016/j.ygyno.2013.05.040.
    1. Chi DS, et al. Improved progression-free and overall survival in advanced ovarian cancer as a result of a change in surgical paradigm. Gynecol. Oncol. 2009;114:26–31. doi: 10.1016/j.ygyno.2009.03.018.
    1. Narasimhulu DM, Khoury-Collado F, Chi DS. Radical surgery in ovarian cancer. Curr. Oncol. Rep. 2015;17:439. doi: 10.1007/s11912-015-0439-z.
    1. Polterauer S, et al. Prognostic value of residual tumor size in patients with epithelial ovarian cancer FIGO stages IIA-IV: analysis of the OVCAD data. Int. J. Gynecol. Cancer. 2012;22:380–385. doi: 10.1097/IGC.0b013e31823de6ae.
    1. Wimberger P. Influence of residual tumor on outcome in ovarian cancer patients with figo stage IV disease. Ann. Sur. Onco.17, 1642–1648 (2010).
    1. Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J. Clin. Oncol. 2002;20:1248–1259. doi: 10.1200/JCO.2002.20.5.1248.
    1. Geomini P, Bremer G, Kruitwagen R, Mol BW. Diagnostic accuracy of frozen section diagnosis of the adnexal mass: a metaanalysis. Gynecol. Oncol. 2005;96:1–9. doi: 10.1016/j.ygyno.2004.09.042.
    1. Ilvan S, et al. The accuracy of frozen section (intraoperative consultation) in the diagnosis of ovarian masses. Gynecol. Oncol. 2005;97:395–399. doi: 10.1016/j.ygyno.2005.01.037.
    1. Maheshwari A, et al. Accuracy of intraoperative frozen section in the diagnosis of ovarian neoplasms: experience at a tertiary oncology center. World J. Surg. Oncol. 2006;4:12. doi: 10.1186/1477-7819-4-12.
    1. Ratnavelu ND, et al. Intraoperative frozen section analysis for the diagnosis of early stage ovarian cancer in suspicious pelvic masses. Cochrane Database Syst. Rev. 2016;3:Cd010360.
    1. Yarandi F, Eftekhar Z, Izadi-Mood N, Shojaei H. Accuracy of intraoperative frozen section in the diagnosis of ovarian tumors. Aust. N. Z. J. Obstet. Gynaecol. 2008;48:438–441. doi: 10.1111/j.1479-828X.2008.00873.x.
    1. Bagade PER, Nayar A. Management of borderline ovarian tumours. Obstet. & Gynaecol. 2012;14:115–120.
    1. Cross PA. Borderline ovarian tumours: a continuing conundrum? BJOG. 2016;123:509. doi: 10.1111/1471-0528.13875.
    1. Schafer KC, et al. In vivo, in situ tissue analysis using rapid evaporative ionization mass spectrometry. Angew. Chem. Int. Ed. Engl. 2009;48:8240–8242. doi: 10.1002/anie.200902546.
    1. Balog J, et al. Intraoperative tissue identification using rapid evaporative ionization mass spectrometry. Sci. Transl. Med. 2013;5:194ra93. doi: 10.1126/scitranslmed.3005623.
    1. Balog J, et al. Identification of biological tissues by rapid evaporative ionization mass spectrometry. Anal. Chem. 2010;82:7343–7350. doi: 10.1021/ac101283x.
    1. Alexander J. et al. (2016) A novel methodology for in vivo endoscopic phenotyping of colorectal cancer based on real-time analysis of the mucosal lipidome: a prospective observational study of the iKnife. Surg. Endosc.31, 1361–1370 (2017).
    1. St John ER, et al. Rapid evaporative ionisation mass spectrometry of electrosurgical vapours for the identification of breast pathology: towards an intelligent knife for breast cancer surgery. Breast Cancer Res. 2017;19:59. doi: 10.1186/s13058-017-0845-2.
    1. Balog J, et al. In vivo endoscopic tissue identification by rapid evaporative ionization mass spectrometry (REIMS) Angew. Chem. Int. Ed. Engl. 2015;54:11059–11062. doi: 10.1002/anie.201502770.
    1. Zhang J. et al. Nondestructive tissue analysis for ex vivo and in vivo cancer diagnosis using a handheld mass spectrometry system. Sci. Transl. Med. 9 (2017), doi: 10.1126/scitranslmed.aan3968
    1. Lengyel E. Ovarian cancer development and metastasis. Am. J. Pathol. 2010;177:1053–1064. doi: 10.2353/ajpath.2010.100105.
    1. Mitra A. K. Ovarian Cancer Metastasis: A Unique Mechanism of Dissemination. Tumor Metastasis, (ed. Xu K.) Ch. 03. (InTech, Rijeka, 2016).
    1. Foster DA. Phosphatidic acid signaling to mTOR: signals for the survival of human cancer cells. Biochim. Biophys. Acta. 2009;1791:949–955. doi: 10.1016/j.bbalip.2009.02.009.
    1. Daly PF, Lyon RC, Faustino PJ, Cohen JS. Phospholipid metabolism in cancer cells monitored by 31P NMR spectroscopy. J. Biol. Chem. 1987;262:14875–14878.
    1. Shan L, et al. Measurement of phospholipids may improve diagnostic accuracy in ovarian cancer. PLoS ONE. 2012;7:e46846. doi: 10.1371/journal.pone.0046846.
    1. Pyragius CE, Fuller M, Ricciardelli C, Oehler MK. Aberrant lipid metabolism: an emerging diagnostic and therapeutic target in ovarian cancer. Int. J. Mol. Sci. 2013;14:7742–7756. doi: 10.3390/ijms14047742.
    1. Sutphen R, et al. Lysophospholipids are potential biomarkers of ovarian cancer. Cancer Epidemiol. Biomark. Prev. 2004;13:1185–1191.
    1. Cai Q, et al. Elevated and secreted phospholipase A(2) activities as new potential therapeutic targets in human epithelial ovarian cancer. FASEB J. 2012;26:3306–3320. doi: 10.1096/fj.12-207597.
    1. Tania M, Khan MA, Song Y. Association of lipid metabolism with ovarian cancer. Curr. Oncol. 2010;17:6–11.
    1. Hu YL, et al. Lysophosphatidic acid induction of vascular endothelial growth factor expression in human ovarian cancer cells. J. Natl. Cancer Inst. 2001;93:762–768. doi: 10.1093/jnci/93.10.762.
    1. Doria ML, et al. Epithelial ovarian carcinoma diagnosis by desorption electrospray ionization mass spectrometry imaging. Sci. Rep. 2016;6:39219. doi: 10.1038/srep39219.

Source: PubMed

3
구독하다