Patient-derived organoids as a predictive biomarker for treatment response in cancer patients

G Emerens Wensink, Sjoerd G Elias, Jasper Mullenders, Miriam Koopman, Sylvia F Boj, Onno W Kranenburg, Jeanine M L Roodhart, G Emerens Wensink, Sjoerd G Elias, Jasper Mullenders, Miriam Koopman, Sylvia F Boj, Onno W Kranenburg, Jeanine M L Roodhart

Abstract

Effective predictive biomarkers are needed to enable personalized medicine and increase treatment efficacy and survival for cancer patients, thereby reducing toxic side effects and treatment costs. Patient-derived organoids (PDOs) enable individualized tumour response testing. Since 2018, 17 publications have examined PDOs as a potential predictive biomarker in the treatment of cancer patients. We review and provide a pooled analysis of the results regarding the use of PDOs in individualized tumour response testing, focusing on evidence for analytical validity, clinical validity and clinical utility. We identify future perspectives to accelerate the implementation of PDOs as a predictive biomarker in the treatment of cancer patients.

Conflict of interest statement

S.F.B. is inventor on patents related to the Organoid Technology. S.F.B. and J.M. are employed by the Foundation Hubrecht Organoid Technology (HUB). The remaining authors declare no conflict of interest. M.K. Institutional financial instructs (IFI): Amgen, Bayer, BMS, Merck-Serono, Nordic Farma, Roche, Servier, Sirtex, Sanofi-Aventis; O.W.K. IFI: Genmab, Johnson & Johnson; J.M.L.R. IFI: Servier, Merck, Bayer. All grants were unrelated to the study and paid to the individual’s institution. All authors are involved in an ongoing prospective clinical trial evaluating the predictive role of organoids in mCRC patients.

Figures

Fig. 1. PDO-based individualized tumour response testing…
Fig. 1. PDO-based individualized tumour response testing as a predictive biomarker.
a Illustrates the tumour types for which patient-derived organoids (PDOs) have been tested for clinical validity (listed in full in Table 1). Personalized treatment strategies currently implemented in oncology treatment largely comprise of genomic biomarkers. However, this only results in a personalized treatment strategy for a minority of patients. Individualized tumour response testing using PDOs is a new biomarker which may be used in personalized treatment and increases access to personalized treatment. b For individualized tumour response testing, tissue from a patient’s tumour is obtained to culture organoids, perform drug screens and various read-outs can be obtained to define PDO drug screen response (including organoid size, viability and co-culture cytokine measurements). A predictive biomarker test is developed using the PDO drug screen results and clinical response seen in patients.
Fig. 2. Evidence landscape of PDO drug…
Fig. 2. Evidence landscape of PDO drug screen parameters and clinical response.
Illustrates the clinical validity results for PDOs as a predictive biomarker for treatment response (dark red: significant correlation and/or predictive value found, pink: trend for correlation or predictive value found, blue: no correlation and white: not tested), with the size of the circle representing the patient cohort size, specified per treatment and tumour type (y-axis) and ex vivo drug response parameter (x-axis). Abbreviations: 5-FU 5-fluorouracil, AC-T doxorubicin + cyclophosphamide + paclitaxel, AUC area under the curve, Capec. capecitabine, CAPIRI capecitabine + irinotecan, CRC colorectal cancer, EOX epirubicin + oxaliplatin + 5-FU, FOLFIRI 5-FU + irinotecan, FOLFOX 5-FU + oxaliplatin, GC gastric cancer, GOC gastroesophageal cancer, GR growth rate inhibition metrics, GR50 value with 50% viable GR, HNSCC head and neck squamous cell carcinoma, ICI immune checkpoint inhibitors, OMI optical metabolic imaging, PDO patient-derived organoid.
Fig. 3. Forest plots of sensitivity and…
Fig. 3. Forest plots of sensitivity and specificity (clinical validity pooled results).
A paired forest plot of the sensitivity and specificity of each study and treatment type is shown with 95% confidence intervals. A bivariate meta-analysis was performed to obtain a pooled summary estimate for sensitivity and specificity indicated in the forest plots (1: for all studies that reported results that could be included in this pooled analysis and 2: for studies with ≥5 responders/non-responders). The analysis was performed in R (Version 3.6.1) using the “mada” package. In blue and bold font (#) the studies were indicated which were included in the analysis for ≥5 responders/non-responders. Patients who contributed to multiple accuracy estimates: 2 patients received ICI and dabrefinib/trametinib; 3 patients received FOLFOX and irinotecan-based treatment; 3 patients had 2 PDOs each (before and after FOLFOX treatment) and 1 patient had a synchronous tumour (responder and non-responder). Abbreviations: capec. capecitabine, CAPIRI capecitabine + irinotecan, df degrees of freedom, EOX epirubicin + oxaliplatin + 5-FU, FOLFIRI 5-flouruoracil + irinotecan, FOLFOX 5-fluorouracil + oxaliplatin, HNSCC head & neck squamous cell carcinoma, ICI immune checkpoint inhibitors, mCRC metastatic colorectal cancer, RC rectal cancer, ref reference, resp. responder clinically, non-resp. non-responder clinically.

References

    1. Ferlay J, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. Eur. J. Cancer. 2018;103:356–387. doi: 10.1016/j.ejca.2018.07.005.
    1. Sawyers CL. The cancer biomarker problem. Nature. 2008;452:548–552. doi: 10.1038/nature06913.
    1. Letai A. Functional precision cancer medicine-moving beyond pure genomics. Nat. Med. 2017;23:1028–1035. doi: 10.1038/nm.4389.
    1. Meric-Bernstam F, et al. Feasibility of large-scale genomic testing to facilitate enrollment onto genomically matched clinical trials. JCO. 2015;33:2753–2762. doi: 10.1200/JCO.2014.60.4165.
    1. van der Velden DL, et al. The Drug Rediscovery protocol facilitates the expanded use of existing anticancer drugs. Nature. 2019;574:127–131. doi: 10.1038/s41586-019-1600-x.
    1. De Souza N. Organoids. Nat. Methods. 2018;15:23. doi: 10.1038/nmeth.4576.
    1. Li M, Izpisua Belmonte JC. Organoids—preclinical models of human disease. NEJM. 2019;380:569–579. doi: 10.1056/NEJMra1806175.
    1. Sato T, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–1772. doi: 10.1053/j.gastro.2011.07.050.
    1. Van De Wetering M, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161:933–945. doi: 10.1016/j.cell.2015.03.053.
    1. Pampaloni F, Reynaud EG, Stelzer EHK. The third dimension bridges the gap between cell culture and live tissue. Nat. Rev. Mol. Cell Biol. 2007;8:839–845. doi: 10.1038/nrm2236.
    1. Vlachogiannis G, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920–926. doi: 10.1126/science.aao2774.
    1. Simon R. Clinical trial designs for evaluating the medical utility of prognostic and predictive biomarkers in oncology. Per Med. 2010;7:33–47. doi: 10.2217/pme.09.49.
    1. Driehuis E, Kretzschmar K, Clevers H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 2020;15:3380–3409. doi: 10.1038/s41596-020-0379-4.
    1. Sachs N, et al. A Living Biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172:373–386.e10. doi: 10.1016/j.cell.2017.11.010.
    1. Kopper O, et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat. Med. 2019;25:838–849. doi: 10.1038/s41591-019-0422-6.
    1. Miao Y, et al. Next-generation surrogate Wnts support organoid growth and deconvolute frizzled pleiotropy in vivo. Cell Stem Cell. 2020;27:840–851.e6. doi: 10.1016/j.stem.2020.07.020.
    1. Sharick JT, et al. Metabolic heterogeneity in patient tumor-derived organoids by primary site and drug treatment. Front. Oncol. 2020;10:1–17. doi: 10.3389/fonc.2020.00553.
    1. Phan N, et al. A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Commun. Biol. 2019;2:1–11. doi: 10.1038/s42003-019-0305-x.
    1. Votanopoulos KI, et al. Model of patient-specific immune-enhanced organoids for immunotherapy screening: feasibility study. Ann. Surg. Oncol. 2019;27:1956–1967. doi: 10.1245/s10434-019-08143-8.
    1. Mazzocchi AR, Rajan SAP, Votanopoulos KI, Hall AR, Skardal A. In vitro patient-derived 3D mesothelioma tumor organoids facilitate patient-centric therapeutic screening. Sci. Rep. 2018;8:1–12. doi: 10.1038/s41598-018-21200-8.
    1. Ooft SN, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci. Transl. Med. 2019;11:eaay2574. doi: 10.1126/scitranslmed.aay2574.
    1. Chalabi M, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat. Med. 2020;26:566–576. doi: 10.1038/s41591-020-0805-8.
    1. Tiriac H, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8:1112–1129. doi: 10.1158/-18-0349.
    1. Fallahi-Sichani M, Honarnejad S, Heiser LM, Gray JW, Sorger PK. Metrics other than potency reveal systematic variation in responses to cancer drugs. Nat. Chem. Biol. 2013;9:708–714. doi: 10.1038/nchembio.1337.
    1. Huang S, Pang L. Comparing statistical methods for quantifying drug sensitivity based on in vitro dose-response assays. ASSAY Drug Dev. Technol. 2012;10:88–96. doi: 10.1089/adt.2011.0388.
    1. Yao Y, et al. Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer. Cell Stem Cell. 2020;26:17–26.e6. doi: 10.1016/j.stem.2019.10.010.
    1. Li X, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat. Commun. 2018;9:2983. doi: 10.1038/s41467-018-05190-9.
    1. Narasimhan V, et al. Medium-throughput drug screening of patient-derived organoids from colorectal peritoneal metastases to direct personalized therapy. CCR. 2020;26:3662–3670.
    1. Ganesh K, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat. Med. 2019;25:1607–1614. doi: 10.1038/s41591-019-0584-2.
    1. Jacob F, et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter- and intra-tumoral heterogeneity. Cell. 2020;180:188–204.e22. doi: 10.1016/j.cell.2019.11.036.
    1. Steele NG, et al. An organoid-based preclinical model of human. Gastric Cancer Cmgh. 2019;7:161–184.
    1. de Witte CJ, et al. Patient-derived ovarian cancer organoids mimic clinical response and exhibit heterogeneous inter- and intrapatient drug responses. Cell Rep. 2020;31:107762. doi: 10.1016/j.celrep.2020.107762.
    1. Hey SP, Gerlach CV, Dunlap G, Prasad V, Kesselheim AS. The evidence landscape in precision medicine. Sci. Transl. Med. 2020;12:1–5. doi: 10.1126/scitranslmed.aaw7745.
    1. Driehuis E, et al. Oral mucosal organoids as a potential platform for personalized cancer therapy. Cancer Discov. 2019;9:852–871. doi: 10.1158/-18-1522.
    1. Varbanov HP, et al. Oxaliplatin reacts with DMSO only in the presence of water. Dalt. Trans. 2017;46:8929–8932. doi: 10.1039/C7DT01628J.
    1. Hinohara K, Polyak K. Intratumoral heterogeneity: more than just mutations. Trends Cell Biol. 2019;29:569–579. doi: 10.1016/j.tcb.2019.03.003.
    1. Bruun J, Kryeziu K, Eide PW, Moosavi SH, Eilertsen IA. Patient-derived organoids from multiple colorectal cancer liver metastases reveal moderate intra-patient pharmacotranscriptomic heterogeneity Translational relevance. CCR. 2020;26:4107–4119.
    1. Verissimo, C. S. et al. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. Elife 1–26 (2016), 10.7554/eLife.18489.
    1. Altman DG, McShane LM, Sauerbrei W, Taube SE. Reporting recommendations for tumor marker prognostic studies (REMARK): explanation and elaboration. BMC Med. 2012;10:1–39. doi: 10.1186/1741-7015-10-51.
    1. Vukicevic S, et al. Identification of multiple active growth factors in basement membrane matrigel suggests caution in interpretation of cellular activity related to extracellular matrix components. Exp. Cell Res. 1992;202:1–8. doi: 10.1016/0014-4827(92)90397-Q.
    1. Hernandez-Gordillo V, et al. Fully synthetic matrices for in vitro culture of primary human intestinal enteroids and endometrial organoids. Biomaterials. 2020;254:120125. doi: 10.1016/j.biomaterials.2020.120125.
    1. Ng S, Tan WJ, Pek MMX, Tan MH, Kurisawa M. Mechanically and chemically defined hydrogel matrices for patient-derived colorectal tumor organoid culture. Biomaterials. 2019;219:119400. doi: 10.1016/j.biomaterials.2019.119400.
    1. Gjorevski N, et al. Designer matrices for intestinal stem cell and organoid culture. Nature. 2016;539:560–564. doi: 10.1038/nature20168.
    1. Sorrentino G, et al. Mechano-modulatory synthetic niches for liver organoid derivation. Nat. Commun. 2020;11:1–10. doi: 10.1038/s41467-020-17161-0.
    1. Schuster B, et al. Automated microfluidic platform for dynamic and combinatorial drug screening of tumor organoids. Nat. Commun. 2020;11:1–12.
    1. Renner H, et al. A fully automated high-throughput workflow for 3D-based chemical screening in human midbrain organoids. Elife. 2020;9:1–39. doi: 10.7554/eLife.52904.
    1. Du Y, et al. Development of a miniaturized 3D organoid culture platform for ultra-high-throughput screening. J. Mol. Cell Biol. 2020;12:630–643. doi: 10.1093/jmcb/mjaa036.
    1. Son B, et al. The role of tumor microenvironment in therapeutic resistance. Oncotarget. 2017;8:3933–3945. doi: 10.18632/oncotarget.13907.
    1. Wilhelm-Benartzi CS, Mt-Isa S, Fiorentino F, Brown R, Ashby D. Challenges and methodology in the incorporation of biomarkers in cancer clinical trials. Crit. Rev. Oncol. Hematol. 2017;110:49–61. doi: 10.1016/j.critrevonc.2016.12.008.
    1. Polley MYC, et al. Statistical and practical considerations for clinical evaluation of predictive biomarkers. J. Natl Cancer Inst. 2013;105:1677–1683. doi: 10.1093/jnci/djt282.
    1. R Core Team. R: A Language and Environment For Statistical Computing (R Foundation for Statistical Computing, 2018).

Source: PubMed

3
Předplatit