Metabolic Heterogeneity in Patient Tumor-Derived Organoids by Primary Site and Drug Treatment

Joe T Sharick, Christine M Walsh, Carley M Sprackling, Cheri A Pasch, Dan L Pham, Karla Esbona, Alka Choudhary, Rebeca Garcia-Valera, Mark E Burkard, Stephanie M McGregor, Kristina A Matkowskyj, Alexander A Parikh, Ingrid M Meszoely, Mark C Kelley, Susan Tsai, Dustin A Deming, Melissa C Skala, Joe T Sharick, Christine M Walsh, Carley M Sprackling, Cheri A Pasch, Dan L Pham, Karla Esbona, Alka Choudhary, Rebeca Garcia-Valera, Mark E Burkard, Stephanie M McGregor, Kristina A Matkowskyj, Alexander A Parikh, Ingrid M Meszoely, Mark C Kelley, Susan Tsai, Dustin A Deming, Melissa C Skala

Abstract

New tools are needed to match cancer patients with effective treatments. Patient-derived organoids offer a high-throughput platform to personalize treatments and discover novel therapies. Currently, methods to evaluate drug response in organoids are limited because they overlook cellular heterogeneity. In this study, non-invasive optical metabolic imaging (OMI) of cellular heterogeneity was characterized in breast cancer (BC) and pancreatic cancer (PC) patient-derived organoids. Baseline heterogeneity was analyzed for each patient, demonstrating that single-cell techniques, such as OMI, are required to capture the complete picture of heterogeneity present in a sample. Treatment-induced changes in heterogeneity were also analyzed, further demonstrating that these measurements greatly complement current techniques that only gauge average cellular response. Finally, OMI of cellular heterogeneity in organoids was evaluated as a predictor of clinical treatment response for the first time. Organoids were treated with the same drugs as the patient's prescribed regimen, and OMI measurements of heterogeneity were compared to patient outcome. OMI distinguished subpopulations of cells with divergent and dynamic responses to treatment in living organoids without the use of labels or dyes. OMI of organoids agreed with long-term therapeutic response in patients. With these capabilities, OMI could serve as a sensitive high-throughput tool to identify optimal therapies for individual patients, and to develop new effective therapies that address cellular heterogeneity in cancer.

Keywords: breast cancer; cellular metabolism; heterogeneity; optical metabolic imaging; organoid; pancreatic cancer.

Copyright © 2020 Sharick, Walsh, Sprackling, Pasch, Pham, Esbona, Choudhary, Garcia-Valera, Burkard, McGregor, Matkowskyj, Parikh, Meszoely, Kelley, Tsai, Deming and Skala.

Figures

Figure 1
Figure 1
Patient-derived tumor organoid generation and optical metabolic imaging. (A) Pie charts depicting the success rate for generating viable organoids from patient pancreatic lesions (left), the distribution of PDAC, PanIN, anaplastic cancer, and ampullary cancer among successfully generated organoids (center), and the distribution of previously treated vs. untreated tumors among successfully generated organoids (right). Representative redox ratio (B), NAD(P)H τm(C), and FAD τm(D) images of an untreated pancreatic organoid taken 6 days after surgical resection (Patient PC14). Scale bar is 50 μm (E). Masks of individual cell cytoplasms overlaid onto NAD(P)H intensity image (F). Representative brightfield image of pancreatic organoids (Patient PC14). Scale bar is 200 μm. (G) Pie charts depicting the success rate for generating viable organoids from patient breast tumor biopsies (left), and the distribution of receptor subtypes among successfully generated organoids (right). Representative redox ratio (H), NAD(P)H τm(I), and FAD τm(J) images of an untreated breast cancer organoid taken 30 days after biopsy (Patient BC9). Scale bar is 50 μm. (K) Masks of individual cell cytoplasms overlaid onto NAD(P)H intensity image. (L) Representative brightfield image of breast cancer organoids (Patient BC9). Scale bar is 200 μm.
Figure 2
Figure 2
Sources of baseline metabolic variability in patient-derived organoid cells. (A) The percentage of total metabolic variation among each breast cancer patient's cells which can be explained by variation at the organoid level for each variable. (B) The percentage of total metabolic variation among each pancreatic patient's cells which can be explained by variation at the organoid level for each variable. (C) The percentage of total metabolic variation among cells from all breast cancer patients which can be explained by variation between patients for each variable. (D) The percentage of total metabolic variation among cells from all pancreatic patients which can be explained by variation between patients for each variable. Untreated organoid cells from the first measurement time point were included in this analysis.
Figure 3
Figure 3
Disparities in metabolism and intra-organoid heterogeneity by morphology and cancer type. (A) Representative brightfield images of hollow breast cancer organoids. Scale bar = 100 μm. (B) Representative NAD(P)H intensity images of hollow breast and pancreatic cancer organoids. Scale bar = 100 μm. (C) Representative brightfield images of solid breast cancer organoids. Scale bar = 250 μm. (D) Representative NAD(P)H intensity images of solid breast and pancreatic cancer organoids. Scale bar = 100 μm. (E–H) The intra-organoid standard deviation of the OMI index (E), coefficient of variation of the redox ratio (F), coefficient of variation of mean fluorescence lifetime of FAD (G), and the mean fluorescence lifetime of NAD(P)H (H) for cells within hollow vs. solid organoids. *p < 0.05. Each dot represents one organoid (mean ± SEM).
Figure 4
Figure 4
OMI captures non-genetic cellular heterogeneity in pancreatic organoids. (A) Representative images of the redox ratio, NAD(P)H τm, and FAD τm, in organoids generated from Patient PC13 (anaplastic carcinoma of the pancreas), treated with standard chemotherapies and experimental targeted therapies for 72 h. Scale bar is 50 μm. (B,C) Heatmap representation of the OMI index treatment effect size (Glass's Δ) for all patients at 72 h in organoids (B) and fibroblasts cultured with organoids. (C) “∧” indicates the patient lesion was diagnosed as PanIN. “~” indicates the patient lesion was diagnosed as ampullary cancer. *Glass's Δ ≥ 0.75. (D) Normalized density distributions of the OMI index of individual cells contain subpopulations with G+P treatment, but not ABT-263 + T treatment after 72 h in PC13 organoids. Bracketed number indicates number of subpopulations. (E) High-depth targeted cancer gene sequencing of PC13 organoids. Allele frequencies of ~50% for KRAS and 100% for TP53 were found (black bars). Alterations with allele frequencies of 10–30% were detected (gray bars), but none of these alterations were pathogenic. (F) Cleaved caspase-3 staining of PC13 organoids shows differences in apoptosis between treatment conditions after 72 h of treatment. (G) Ki67 staining of PC13 organoids shows differences in proliferation between treatment conditions after 72 h. Each dot represents one organoid (mean ± SEM), and red indicates significant response to treatment. *p < 0.05 vs. control.
Figure 5
Figure 5
Organoid-based predictions of pancreatic cancer patient response to therapy. (A–D) Representative redox ratio, NAD(P)H τm, and FAD τm images of pancreatic organoids from Patients PC1 (A), PC2 (B), PC6 (C), and PC14 (D), who are classified as predicted responders. Left columns indicate control organoids, and right columns indicate organoids treated with the same drugs as the patient adjuvant treatment. Scale bar is 50 μm. (E–H) The effect of the same drugs on the OMI index averaged across all cells in organoids derived from Patient PC1 (E), PC2 (F), PC6 (G), and PC14 (H). Error bars indicate mean ± SEM. *p < 0.0001. (I–L) Single-cell OMI index subpopulation analysis of treatment response in organoids from Patient PCI (I), PC2 (J), PC6 (K), and PC14 (L). (M–O) Representative redox ratio, NAD(P)H τm, and FAD τm images of pancreatic organoids from Patients PC3 (M), PC8 (N), and PC18 (O), who are classified as predicted non-responders. Left columns indicate control organoids, and right columns indicate organoids treated with the same drugs as the patient adjuvant treatment. (P–R) The effect of the same drugs on the OMI index averaged across all cells in organoids derived from Patient PC3 (P), PC8 (Q), and PC18 (R). (S–U) Single-cell OMI index subpopulation analysis of treatment response in organoids from Patient PC3 (S), PC8 (T), and PC18 (U).
Figure 6
Figure 6
Pancreatic cancer patient clinical outcomes while on adjuvant therapy. (A) Swimmer plot indicating the number of months without recurrence following surgical resection of the tumor and adjuvant treatment. Patients are classified as predicted responders and non-responders based on organoid response profiles. Arrows indicate that the patient continues to survive without recurrence at the time of publication. “~” Indicates the patient's lesion was diagnosed as ampullary cancer. (B) Patients with RFS > 12 months had higher OMI index effect sizes at 72 h (Glass's Δ) than patients with RFS <12 months (mean± SEM). Dotted line represents proposed cutoff of Δ = 0.75. (C) Patients with RFS > 12 months show a decrease in wH-index with treatment compared to control organoids. Patients with RFS < 12 months show an increase in wH-index with treatment compared here to control organoids. Error bars not visible. N = 1,000 fits/group.
Figure 7
Figure 7
OMI of single-cell treatment response in patient-derived breast tumor organoids. (A) Representative images of the optical redox ratio, NAD(P)H τm, and FAD τm organoids generated from Patient BC8 after 72 h of treatment. Scale bar = 100 μm. (B) Heatmap representation of the OMI index treatment effect size (Glass's Δ) at 72 h in organoids from each breast cancer patient. *Glass's Δ ≥ 0.75. 4-OOH cyclophosphamide (active metabolite) was used in place of cyclophosphamide. (C,D) Representative images of control (C) and A+C+T treated (D) BC8 organoids stained for Ki67 (green, proliferation), cleaved caspase-3 (red, apoptosis), and DAPI (blue, nuclei) after 72 h of treatment. Scale bar 100 μm. (E) Cleaved caspase-3 staining of organoids shows differences in apoptosis between treatment conditions after 72 h of treatment in BC8. (F) Ki67 staining of organoids shows differences in proliferation between treatment conditions after 72 h in BC8. Each dot represents one organoid (mean ± SEM). *p < 0.05 vs. control. (G) The effect of A+C+T treatment at 72 h on OMI index heterogeneity quantified by the wH-index in Patient BC8, BC17, and BC20 organoids. (H–J) The effect of 72 h A+C+T treatment on the OMI index averaged across all cells in organoids derived from Patient BC8 (H), BC17 (I), and BC20 (J). Error bars indicate mean ± SEM. *p < 0.0001. Single cell OMI index subpopulation analysis of 72 h of A+C+T treatment response in organoids from Patient BC8 (K), BC17 (L), and BC20 (M).

References

    1. Walsh AJ, Castellanos JA, Nagathihalli NS, Merchant NB, Skala MC. Optical imaging of drug-induced metabolism changes in murine and human pancreatic cancer organoids reveals heterogeneous drug response. Pancreas. (2016) 45:863–9. 10.1097/MPA.0000000000000543
    1. Moreira L, Bakir B, Chatterji P, Dantes Z, Reichert M, Rustgi AK. Pancreas 3D organoids: current and future aspects as a research platform for personalized medicine in pancreatic cancer. Cell Mol Gastroenterol Hepatol. (2018) 5:289–98. 10.1016/j.jcmgh.2017.12.004
    1. Wolff RA, Wang-Gillam A, Alvarez H, Tiriac H, Engle D, Hou S, et al. . Dynamic changes during the treatment of pancreatic cancer. Oncotarget. (2018) 9:14764–90. 10.18632/oncotarget.24483
    1. Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, et al. . Organoid models of human and mouse ductal pancreatic cancer. Cell. (2015) 160:324–38. 10.1016/j.cell.2014.12.021
    1. Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville T, et al. . Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. (2018) 10.1158/1538-7445.PANCA19-C57
    1. Huang L, Holtzinger A, Jagan I, BeGora M, Lohse I, Ngai N, et al. . Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids. Nat Med. (2015) 21:1364–71. 10.1038/nm.3973
    1. Romero-Calvo I, Weber C, Ray M, Brown M, Kirby K, Nandi RK, et al. . Human organoids share structural and genetic features with primary pancreatic adenocarcinoma tumors. Mol Cancer Res. (2018) 17:70-83. 10.1158/1541-7786.MCR-18-0531
    1. Tsai S, McOlash L, Palen K, Johnson B, Duris C, Yang Q, et al. . Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer. (2018) 18:335. 10.1186/s12885-018-4238-4
    1. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. . A living biobank of breast cancer organoids captures disease heterogeneity. Cell. (2018) 172:373–86.e10. 10.1016/j.cell.2017.11.010
    1. Jenkins RW, Aref AR, Lizotte PH, Ivanova E, Stinson S, Zhou CW, et al. . Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. (2018) 8:196–215. 10.1158/-17-0833
    1. Majety M, Pradel LP, Gies M, Ries CH. Fibroblasts influence survival and therapeutic response in a 3D co-culture model. PLoS ONE. (2015) 10:e0127948. 10.1371/journal.pone.0127948
    1. Micke P, Ostman A. Tumour-stroma interaction: cancer-associated fibroblasts as novel targets in anti-cancer therapy? Lung Cancer. (2004) 45(Suppl. 2):S163–75. 10.1016/j.lungcan.2004.07.977
    1. Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. (2013) 501:346–54. 10.1038/nature12626
    1. Marusyk A, Polyak K. Tumor heterogeneity: causes and consequences. Biochim Biophys Acta. (2010) 1805:105–17. 10.1016/j.bbcan.2009.11.002
    1. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. (2013) 501:338–45. 10.1038/nature12625
    1. Gerlinger M, Swanton C. How darwinian models inform therapeutic failure initiated by clonal heterogeneity in cancer medicine. Br J Cancer. (2010) 103:1139–43. 10.1038/sj.bjc.6605912
    1. Fisher R, Pusztai L, Swanton C. Cancer heterogeneity: implications for targeted therapeutics. Br J Cancer. (2013) 108:479–85. 10.1038/bjc.2012.581
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. (2011) 144:646–74. 10.1016/j.cell.2011.02.013
    1. Rahman M, Hasan MR. Cancer metabolism and drug resistance. Metabolites. (2015) 5:571–600. 10.3390/metabo5040571
    1. Cantor JR, Sabatini DM. Cancer cell metabolism: one hallmark, many faces. Cancer Discov. (2012) 2:881–98. 10.1158/-12-0345
    1. Shi X, Tasdogan A, Huang F, Hu Z, Morrison SJ, DeBerardinis RJ. The abundance of metabolites related to protein methylation correlates with the metastatic capacity of human melanoma xenografts. Sci Adv. (2017) 3:eaao5268. 10.1126/sciadv.aao5268
    1. Skala MC, Riching KM, Gendron-Fitzpatrick A, Eickhoff J, Eliceiri KW, White JG, et al. . In vivo multiphoton microscopy of NADH and FAD redox states, fluorescence lifetimes, and cellular morphology in precancerous epithelia. Proc Natl Acad Sci USA. (2007) 104:19494–9. 10.1073/pnas.0708425104
    1. Walsh AJ, Cook RS, Manning HC, Hicks DJ, Lafontant A, Arteaga CL, et al. . Optical metabolic imaging identifies glycolytic levels, subtypes, and early-treatment response in breast cancer. Cancer Res. (2013) 73:6164–74. 10.1158/0008-5472.CAN-13-0527
    1. Georgakoudi I, Quinn KP. Optical imaging using endogenous contrast to assess metabolic state. Annu Rev Biomed Eng. (2012) 14:351–67. 10.1146/annurev-bioeng-071811-150108
    1. Chance B, Schoener B, Oshino R, Itshak F, Nakase Y. Oxidation-reduction ratio studies of mitochondria in freeze-trapped samples. NADH and flavoprotein fluorescence signals. J Biol Chem. (1979) 254:4764–71.
    1. Walsh A, Cook RS, Rexer B, Arteaga CL, Skala MC. Optical imaging of metabolism in HER2 overexpressing breast cancer cells. Biomed Opt Express. (2012) 3:75–85. 10.1364/BOE.3.000075
    1. Alhallak K, Rebello LG, Muldoon TJ, Quinn KP, Rajaram N. Optical redox ratio identifies metastatic potential-dependent changes in breast cancer cell metabolism. Biomed Opt Express. (2016) 7:4364–74. 10.1364/BOE.7.004364
    1. Hou J, Wright HJ, Chan N, Tran R, Razorenova OV, Potma EO, et al. . Correlating two-photon excited fluorescence imaging of breast cancer cellular redox state with seahorse flux analysis of normalized cellular oxygen consumption. J Biomed Opt. (2016) 21:60503. 10.1117/1.JBO.21.6.060503
    1. Sharick JT, Favreau PF, Gillette AA, Sdao SM, Merrins MJ, Skala MC. Protein-bound NAD(P)H lifetime is sensitive to multiple fates of glucose carbon. Sci Rep. (2018) 8:5456. 10.1038/s41598-018-23691-x
    1. Lakowicz JR. Principles of Fluorescence Spectroscopy. Vol. 23 2nd ed New York, NY: Kluwer Academic/Plenum; (1999). p. 698 10.1007/978-1-4757-3061-6_19
    1. Bird DK, Yan L, Vrotsos KM, Eliceiri KW, Vaughan EM, Keely PJ, et al. . Metabolic mapping of MCF10A human breast cells via multiphoton fluorescence lifetime imaging of the coenzyme NADH. Cancer Res. (2005) 65:8766–73. 10.1158/0008-5472.CAN-04-3922
    1. Alam SR, Wallrabe H, Svindrych Z, Chaudhary AK, Christopher KG, Chandra D, et al. . Investigation of mitochondrial metabolic response to doxorubicin in prostate cancer cells: an NADH, FAD and tryptophan FLIM assay. Sci Rep. (2017) 7:10451. 10.1038/s41598-017-10856-3
    1. Lukina MM, Dudenkova VV, Ignatova NI, Druzhkova IN, Shimolina LE, Zagaynova EV, et al. . Metabolic cofactors NAD(P)H and FAD as potential indicators of cancer cell response to chemotherapy with paclitaxel. Biochim Biophys Acta Gen Sub. (2018) 1862:1693–700. 10.1016/j.bbagen.2018.04.021
    1. Wallrabe H, Svindrych Z, Alam SR, Siller KH, Wang T, Kashatus D, et al. . Segmented cell analyses to measure redox states of autofluorescent NAD(P)H, FAD & Trp in cancer cells by FLIM. Sci Rep. (2018) 8:79. 10.1038/s41598-017-18634-x
    1. Walsh AJ, Cook RS, Sanders ME, Aurisicchio L, Ciliberto G, Arteaga CL, et al. . Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res. (2014) 74:5184–94. 10.1158/0008-5472.CAN-14-0663
    1. Shah AT, Diggins KE, Walsh AJ, Irish JM, Skala MC. In vivo autofluorescence imaging of tumor heterogeneity in response to treatment. Neoplasia. (2015) 17:862–70. 10.1016/j.neo.2015.11.006
    1. Walsh AJ, Skala MC. Optical metabolic imaging quantifies heterogeneous cell populations. Biomed Opt Express. (2015) 6:559–73. 10.1364/BOE.6.000559
    1. Pasch CA, Favreau PF, Yueh AE, Babiarz CP, Gillette AA, Sharick JT, et al. . Patient-derived cancer organoid cultures to predict sensitivity to chemotherapy and radiation. Clin Cancer Res. (2019) 25:5376-538. 10.1158/1078-0432.CCR-18-3590
    1. Witkiewicz AK, Balaji U, Eslinger C, McMillan E, Conway W, Posner B, et al. . Integrated patient-derived models delineate individualized therapeutic vulnerabilities of pancreatic cancer. Cell Rep. (2016) 16:2017–31. 10.1016/j.celrep.2016.07.023
    1. Shibata T, Ebata T, Fujita K, Shimokata T, Maeda O, Mitsuma A, et al. Optimal dose of gemcitabine for the treatment of biliary tract or pancreatic cancer in patients with liver dysfunction. Cancer Sci. (2016) 107:168–72. 10.1111/cas.12851
    1. Kroep JR, Giaccone G, Voorn DA, Smit EF, Beijnen JH, Rosing H, et al. . Gemcitabine and paclitaxel: pharmacokinetic and pharmacodynamic interactions in patients with non-small-cell lung cancer. J Clin Oncol. (1999) 17:2190–7. 10.1200/JCO.1999.17.7.2190
    1. Huang L, Bockorny B, Paul I, Akshinthala D, Gandarilla O, Bose A, et al. Pancreatic tumor organoids for modeling in vivo drug response and discovering clinically-actionable biomarkers. bioRxiv. (2019) 2019:513267 10.1101/513267
    1. Jodrell DI, Stewart M, Aird R, Knowles G, Bowman A, Wall L, et al. . 5-fluorouracil steady state pharmacokinetics and outcome in patients receiving protracted venous infusion for advanced colorectal cancer. Br J Cancer. (2001) 84:600–3. 10.1054/bjoc.2000.1664
    1. Garcia-Garcia C, Ibrahim YH, Serra V, Calvo MT, Guzman M, Grueso J, et al. . Dual mTORC1/2 and HER2 blockade results in antitumor activity in preclinical models of breast cancer resistant to anti-HER2 therapy. Clin Cancer Res. (2012) 18:2603–12. 10.1158/1078-0432.CCR-11-2750
    1. Infante JR, Tabernero J, Cervantes A, Jalal S, Burris HA, Macarulla T, et al. Abstract C252: a phase 1, dose-escalation study of MLN0128, an investigational oral mammalian target of rapamycin complex 1/2 (mTORC1/2) catalytic inhibitor, in patients (pts) with advanced non-hematologic malignancies. Mol Cancer Ther. (2013) 12(Suppl. 11):C252 10.1158/1535-7163.TARG-13-C252
    1. Gandhi L, Camidge DR, de Oliveira MR, Bonomi P, Gandara D, Khaira D, et al. . Phase I study of navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors. J Clin Oncol. (2011) 29:909–16. 10.1200/JCO.2010.31.6208
    1. Shoemaker AR, Mitten MJ, Adickes J, Ackler S, Refici M, Ferguson D, et al. . Activity of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models. Clin Cancer Res. (2008) 14:3268–77. 10.1158/1078-0432.CCR-07-4622
    1. Graham MA, Lockwood GF, Greenslade D, Brienza S, Bayssas M, Gamelin E. Clinical pharmacokinetics of oxaliplatin: a critical review. Clin Cancer Res. (2000) 6:1205–18.
    1. Mathijssen RH, van Alphen RJ, Verweij J, Loos WJ, Nooter K, Stoter G, et al. . Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res. (2001) 7:2182–94.
    1. Baker SD, Zhao M, Lee CK, Verweij J, Zabelina Y, Brahmer JR, et al. . Comparative pharmacokinetics of weekly and every-three-weeks docetaxel. Clin Cancer Res. (2004) 10:1976–83. 10.1158/1078-0432.CCR-0842-03
    1. Joy MS, La M, Wang J, Bridges AS, Hu Y, Hogan SL, et al. . Cyclophosphamide and 4-hydroxycyclophosphamide pharmacokinetics in patients with glomerulonephritis secondary to lupus and small vessel vasculitis. Br J Clin Pharmacol. (2012) 74:445–55. 10.1111/j.1365-2125.2012.04223.x
    1. Tian YF, Ahn H, Schneider RS, Yang SN, Roman-Gonzalez L, Melnick AM, et al. . Integrin-specific hydrogels as adaptable tumor organoids for malignant B and T cells. Biomaterials. (2015) 73:110–9. 10.1016/j.biomaterials.2015.09.007
    1. Pickl M, Ries CH. Comparison of 3D and 2D tumor models reveals enhanced HER2 activation in 3D associated with an increased response to trastuzumab. Oncogene. (2009) 28:461–8. 10.1038/onc.2008.394
    1. Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res Treat. (2010) 122:35–43. 10.1007/s10549-009-0502-2
    1. Bergmann A. SPCImage: Data Analysis Software for Fluorescence Lifetime Imaging Microscopy. Becker & Hickl GmbH (2003). Available online at: (accessed November 15, 2018).
    1. Lakowicz JR, Szmacinski H, Nowaczyk K, Johnson ML. Fluorescence lifetime imaging of free and protein-bound NADH. Proc Natl Acad Sci USA. (1992) 89:1271–5. 10.1073/pnas.89.4.1271
    1. Nakashima N, Yoshihara K, Tanaka F, Yagi K. Picosecond fluorescence lifetime of the coenzyme of D-amino acid oxidase. J Biol Chem. (1980)255:5261–3.
    1. Walsh AJ, Skala MC. An automated image processing routine for segmentation of cell cytoplasms in high-resolution autofluorescence images. SPIE Proc. (2014) 14:8948 10.1117/12.2040644
    1. Carpenter AE, Jones TR, Lamprecht MR, Clarke C, Kang IH, Friman O, et al. . CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. (2006) 7:R100. 10.1186/gb-2006-7-10-r100
    1. Pan W, Lin J, Le CT. Model-based cluster analysis of microarray gene-expression data. Genome Biol. (2002) 3:research0009.1. 10.1186/gb-2002-3-2-research0009
    1. Sharick JT, Jeffery JJ, Karim MR, Walsh CM, Esbona K, Cook RS, et al. . Cellular metabolic heterogeneity in vivo is recapitulated in tumor organoids. Neoplasia. (2019) 21:615–26. 10.1016/j.neo.2019.04.004
    1. Akaike H. A new look at the statistical model identification. IEEE Trans Automatic Control. (1974) 19:716–23. 10.1109/TAC.1974.1100705
    1. Almendro V, Kim HJ, Cheng YK, Gonen M, Itzkovitz S, Argani P, et al. . Genetic and phenotypic diversity in breast tumor metastases. Cancer Res. (2014) 74:1338–48. 10.1158/0008-5472.CAN-13-2357-T
    1. Gough AH, Chen N, Shun TY, Lezon TR, Boltz RC, Reese CE, et al. . Identifying and quantifying heterogeneity in high content analysis: application of heterogeneity indices to drug discovery. PLoS ONE. (2014) 9:e102678. 10.1371/journal.pone.0102678
    1. Glass GV. Primary, secondary, and meta-analysis of research. Educ Res. (1976) 5:3–8. 10.3102/0013189X005010003
    1. Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. . Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. (2017) 214:579–96. 10.1084/jem.20162024
    1. Sievers CK, Zou LS, Pickhardt PJ, Matkowskyj KA, Albrecht DM, Clipson L, et al. . Subclonal diversity arises early even in small colorectal tumours and contributes to differential growth fates. Gut. (2017) 66:2132–40. 10.1136/gutjnl-2016-312232
    1. Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. . Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol. (2007) 25:4414–22. 10.1200/JCO.2007.10.6823
    1. O'Brien LE, Zegers MM, Mostov KE. Opinion: building epithelial architecture: insights from three-dimensional culture models. Nat Rev Mol Cell Biol. (2002) 3:531–7. 10.1038/nrm859
    1. Zanoni M, Piccinini F, Arienti C, Zamagni A, Santi S, Polico R, et al. . 3D tumor spheroid models for in vitro therapeutic screening: a systematic approach to enhance the biological relevance of data obtained. Sci Rep. (2016) 6:19103. 10.1038/srep19103
    1. Weaver VM, Lelievre S, Lakins JN, Chrenek MA, Jones JC, Giancotti F, et al. . beta4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium. Cancer Cell. (2002) 2:205–16. 10.1016/S1535-6108(02)00125-3
    1. Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. (2014) 371:2140–1. 10.1056/NEJMra1404198
    1. Nagathihalli NS, Castellanos JA, Shi C, Beesetty Y, Reyzer ML, Caprioli R, et al. . Signal transducer and activator of transcription 3, mediated remodeling of the tumor microenvironment results in enhanced tumor drug delivery in a mouse model of pancreatic cancer. Gastroenterology. (2015) 149:1932–43.e9. 10.1053/j.gastro.2015.07.058
    1. Sharick JT, Walsh CM, Sprackling CM, Pasch CA, Parikh AA, Matkowskyj KA, et al. Optical metabolic imaging of heterogeneous drug response in pancreatic cancer patient organoids. bioRxiv. (2019) 2019:542167 10.1101/542167

Source: PubMed

3
Subscribe