Image-based ex-vivo drug screening for patients with aggressive haematological malignancies: interim results from a single-arm, open-label, pilot study

Berend Snijder, Gregory I Vladimer, Nikolaus Krall, Katsuhiro Miura, Ann-Sofie Schmolke, Christoph Kornauth, Oscar Lopez de la Fuente, Hye-Soo Choi, Emiel van der Kouwe, Sinan Gültekin, Lukas Kazianka, Johannes W Bigenzahn, Gregor Hoermann, Nicole Prutsch, Olaf Merkel, Anna Ringler, Monika Sabler, Georg Jeryczynski, Marius E Mayerhoefer, Ingrid Simonitsch-Klupp, Katharina Ocko, Franz Felberbauer, Leonhard Müllauer, Gerald W Prager, Belgin Korkmaz, Lukas Kenner, Wolfgang R Sperr, Robert Kralovics, Heinz Gisslinger, Peter Valent, Stefan Kubicek, Ulrich Jäger, Philipp B Staber, Giulio Superti-Furga, Berend Snijder, Gregory I Vladimer, Nikolaus Krall, Katsuhiro Miura, Ann-Sofie Schmolke, Christoph Kornauth, Oscar Lopez de la Fuente, Hye-Soo Choi, Emiel van der Kouwe, Sinan Gültekin, Lukas Kazianka, Johannes W Bigenzahn, Gregor Hoermann, Nicole Prutsch, Olaf Merkel, Anna Ringler, Monika Sabler, Georg Jeryczynski, Marius E Mayerhoefer, Ingrid Simonitsch-Klupp, Katharina Ocko, Franz Felberbauer, Leonhard Müllauer, Gerald W Prager, Belgin Korkmaz, Lukas Kenner, Wolfgang R Sperr, Robert Kralovics, Heinz Gisslinger, Peter Valent, Stefan Kubicek, Ulrich Jäger, Philipp B Staber, Giulio Superti-Furga

Abstract

Background: Patients with refractory or relapsed haematological malignancies have few treatment options and short survival times. Identification of effective therapies with genomic-based precision medicine is hampered by intratumour heterogeneity and incomplete understanding of the contribution of various mutations within specific cancer phenotypes. Ex-vivo drug-response profiling in patient biopsies might aid effective treatment identification; however, proof of its clinical utility is limited.

Methods: We investigated the feasibility and clinical impact of multiparametric, single-cell, drug-response profiling in patient biopsies by immunofluorescence, automated microscopy, and image analysis, an approach we call pharmacoscopy. First, the ability of pharmacoscopy to separate responders from non-responders was evaluated retrospectively for a cohort of 20 newly diagnosed and previously untreated patients with acute myeloid leukaemia. Next, 48 patients with aggressive haematological malignancies were prospectively evaluated for pharmacoscopy-guided treatment, of whom 17 could receive the treatment. The primary endpoint was progression-free survival in pharmacoscopy-treated patients, as compared with their own progression-free survival for the most recent regimen on which they had progressive disease. This trial is ongoing and registered with ClinicalTrials.gov, number NCT03096821.

Findings: Pharmacoscopy retrospectively predicted the clinical response of 20 acute myeloid leukaemia patients to initial therapy with 88·1% accuracy. In this interim analysis, 15 (88%) of 17 patients receiving pharmacoscopy-guided treatment had an overall response compared with four (24%) of 17 patients with their most recent regimen (odds ratio 24·38 [95% CI 3·99-125·4], p=0·0013). 12 (71%) of 17 patients had a progression-free survival ratio of 1·3 or higher, and median progression-free survival increased by four times, from 5·7 (95% CI 4·1-12·1) weeks to 22·6 (7·4-34·0) weeks (hazard ratio 3·14 [95% CI 1·37-7·22], p=0·0075).

Interpretation: Routine clinical integration of pharmacoscopy for treatment selection is technically feasible, and led to improved treatment of patients with aggressive refractory haematological malignancies in an initial patient cohort, warranting further investigation.

Funding: Austrian Academy of Sciences; European Research Council; Austrian Science Fund; Austrian Federal Ministry of Science, Research and Economy; National Foundation for Research, Technology and Development; Anniversary Fund of the Austrian National Bank; MPN Research Foundation; European Molecular Biology Organization; and Swiss National Science Foundation.

Copyright © 2017 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Pharmacoscopy and response to first-line acute myeloid leukaemia therapy (A) Schematic overview of the retrospective analysis using biobanked bone marrow samples of 20 patients with acute myeloid leukaemia taken before first-line treatment. (B) Comparison of two cell-death readouts: count of cells without activated caspase-3 and count of non-fragmented nuclei. Dots represent values from individual wells, for drug-screen results aggregated from three different patient samples. (C) Heatmap of the DMSO-relative fraction of CD34+ CD117+ blasts (C), and total cell number relative to DMSO (E), averaged for the non-responders and complete remission patient groups. Comparison of the DMSO-relative fraction of CD34+ KIT+ blasts (D) and total cell number (F) for the non-responders and complete remission groups, plotted as function of increasing concentrations of daunorubicin. Data are mean (SE) of patients. p values are for one-sided t test testing for reductions in complete remission group compared to non-responders. (G) Surface plot indicating the ideal separating hyperplane between the complete remission and non-responders groups. The drug space of 25 columns and five rows represents the same drug space as shown in (C) and (E). (H) Integrated RBF response scores per patient. Boxplots show distributions, dots are values for individual patients; crosses indicate datapoints that do not fall between the whiskers. (I) Cross-validation accuracies for integrated response scores for different cellular drug response readouts. (J) Average ROC curves over all cross-validation runs for different cell death readouts. AUROC values are indicated. (C–F) Assays done in technical quadruplicates for each of the 20 patient bone marrow samples. (I–J) Averaged classification results over 2025 cross-validation runs. AUROC=average area under ROC curve. DMSO=dimethyl sulfoxide. ROC=receiver operating characteristics.
Figure 2
Figure 2
Study outline and representative responses to pharmacoscopy-guided treatments (A) Outline of the workflow of this study, including patient numbers (n) and reasons for patient dropping out of the study. (B) Comparison of percentage of marker-positive cells identified by flow cytometry-based diagnostic pathology in the clinic and by automated microscopy and single-cell image analysis in patients for which both datasets were available. (C–F) RBFs for all screened drugs for selected case studies of patients, with corresponding patient numbers, diagnoses, and used blast marker indicated. Bar graphs show drugs ranked by average RBF, indicating significant (dark grey) and non-significant (light grey) RBFs; values of RBF>1·2 are capped at 1·2. Horizontal line at value 1 indicates DMSO-control levels. Table showing top-hit drug names, ranks, RBF, and p values (two-tailed t test against controls); yellow highlights indicate selected drugs provided during pharmacoscopy-guided treatment. (G–J) PET-CT or PET-MRT scans corresponding to the patients described in (A–D), at the time of last relapse (before) and after pharmacoscopy-guided treatments (after). White outlined boxes indicate tumour foci. NS=not significant. RBF=relative blast fractions.
Figure 3
Figure 3
Overall response and progression-free survival with pharmacoscopy-guided treatment (A) Comparison of overall response with the most recent regimen and of pharmacoscopy-guided treatments for 17 patients with aggressive haematological malignancies. p value was calculated by McNemar's test for paired binomial data. (B) Kaplan-Meier plot showing progression-free survival with the most recent regimen and pharmacoscopy-guided treatments for 17 patients. (C) Progression-free survival with most recent regimen or pharmacoscopy-guided treatment per patient. *Ongoing response at time of analysis.
Figure 4
Figure 4
Pharmacoscopy and therapeutic response (A) Average pharmacoscopy scores in all patients per best overall response reveal negative scores associated with progressive disease. (B) i-PCY score per patient by best overall response in 29 patients. Individual dots correspond to individual patients. Bars show average i-PCY scores by overall response. Box and whisker plots show i-PCY scores for progressive disease and partial response and complete remission responses. Inset shows the corresponding ROC curve. Coloured boxes show pharmacoscopy data for all markers and drugs for selected patient; heat map colours range from dark red (iPCY1), see also the legend in the appendix (p 5). i-PCY=integrated pharmacoscopy. (C) Average pharmacoscopy scores. p values directly above bars indicate significant deviation from 0; p values of pairwise comparisons are indicated by corresponding connecting lines (A, C). (D) Box and whisker plots of the tested drugs to which ex-vivo resistance is observed by pharmacoscopy per number of previous treatment lines in 29 patients. Individual patient values are plotted as black dots next to the boxplots. ROC=receiver operating characteristics. Crosses indicate datapoints that do not fall between the whiskers.

References

    1. Khwaja A, Bjorkholm M, Gale RE. Acute myeloid leukaemia. Nat Rev Dis Primers. 2016;2:16010.
    1. Hood L, Friend SH. Predictive, personalized, preventive, participatory (P4) cancer medicine. Nat Rev Clin Oncol. 2011;8:184–187.
    1. Tsimberidou AM, Wen S, Hong DS. Personalized medicine for patients with advanced cancer in the phase I program at MD Anderson: validation and landmark analyses. Clin Cancer Res. 2014;20:4827–4836.
    1. Von Hoff DD, Stephenson JJ, Jr, Rosen P. Pilot study using molecular profiling of patients' tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol. 2010;28:4877–4883.
    1. Le Tourneau C, Delord JP, Goncalves A. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–1334.
    1. Friedman AA, Letai A, Fisher DE, Flaherty KT. Precision medicine for cancer with next-generation functional diagnostics. Nat Rev Cancer. 2015;15:747–756.
    1. Hou HA, Lin CC, Chou WC. Integration of cytogenetic and molecular alterations in risk stratification of 318 patients with de novo non-M3 acute myeloid leukemia. Leukemia. 2014;28:50–58.
    1. Byrd JC, Mrozek K, Dodge RK. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461) Blood. 2002;100:4325–4336.
    1. Schlenk RF, Dohner K, Krauter J. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1909–1918.
    1. Pastore F, Dufour A, Benthaus T. Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2014;32:1586–1594.
    1. Walter RB, Othus M, Paietta EM. Effect of genetic profiling on prediction of therapeutic resistance and survival in adult acute myeloid leukemia. Leukemia. 2015;29:2104–2107.
    1. Zuber J, Radtke I, Pardee TS. Mouse models of human AML accurately predict chemotherapy response. Genes Dev. 2009;23:877–889.
    1. Pfreundschuh M, Trümper L, Osterborg A. CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial (MInT) Group. Lancet Oncol. 2006;7:379–391.
    1. Irish JM, Hovland R, Krutzik PO. Single cell profiling of potentiated phospho-protein networks in cancer cells. Cell. 2004;118:217–228.
    1. Kornblau SM, Minden MD, Rosen DB. Dynamic single-cell network profiles in acute myelogenous leukemia are associated with patient response to standard induction therapy. Clin Cancer Res. 2010;16:3721–3733.
    1. Lacayo NJ, Alonzo TA, Gayko U. Development and validation of a single-cell network profiling assay-based classifier to predict response to induction therapy in paediatric patients with de novo acute myeloid leukaemia: a report from the Children's Oncology Group. Br J Haematol. 2013;162:250–262.
    1. Montero J, Sarosiek KA, DeAngelo JD. Drug-induced death signaling strategy rapidly predicts cancer response to chemotherapy. Cell. 2015;160:977–989.
    1. Frismantas V, Dobay MP, Rinaldi A. Ex vivo drug response profiling detects recurrent sensitivity patterns in drug-resistant acute lymphoblastic leukemia. Blood. 2017;129:e26–e37.
    1. Kurtz SE, Eide CA, Kaempf A. Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic malignancies. Proc Natl Acad Sci USA. 2017;114:e7554–e7563.
    1. Pemovska T, Kontro M, Yadav B. Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia. Cancer Discov. 2013;3:1416–1429.
    1. Majumder MM, Silvennoinen R, Anttila P. Identification of precision treatment strategies for relapsed/refractory multiple myeloma by functional drug sensitivity testing. Oncotarget. 2017;8:56338–56350.
    1. Yu M, Bardia A, Aceto N. Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science. 2014;345:216–220.
    1. Hidalgo M, Amant F, Biankin AV. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4:998–1013.
    1. Roife D, Dai B, Kang Y. Ex vivo testing of patient-derived xenografts mirrors the clinical outcome of patients with pancreatic ductal adenocarcinoma. Clin Cancer Res. 2016;22:6021–6030.
    1. Gao D, Vela I, Sboner A. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159:176–187.
    1. van de Wetering M, Francies HE, Francis JM. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161:933–945.
    1. Selby P, Buick RN, Tannock I. A critical appraisal of the “human tumor stem-cell assay”. N Engl J Med. 1983;308:129–134.
    1. Von Hoff DD. “Send this patient's tumor for culture and sensitivity”. N Engl J Med. 1983;308:154–155.
    1. Samson DJ, Seidenfeld J, Ziegler K, Aronson N. Chemotherapy sensitivity and resistance assays: a systematic review. J Clin Oncol. 2004;22:3618–3630.
    1. Vladimer GI, Snijder B, Krall N. Global survey of the immunomodulatory potential of common drugs. Nat Chem Biol. 2017;13:681–690.
    1. Walter RB, Kantarjian HM, Huang X. Effect of complete remission and responses less than complete remission on survival in acute myeloid leukemia: a combined Eastern Cooperative Oncology Group, Southwest Oncology Group, and M D Anderson Cancer Center Study. J Clin Oncol. 2010;28:1766–1771.
    1. Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 2011;29:487–494.
    1. Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood. 2016;127:53–61.
    1. Wells SJ, Bray RA, Stempora LL, Farhi DC. CD117/CD34 expression in leukemic blasts. Am J Clin Pathol. 1996;106:192–195.
    1. Cheson BD, Pfistner B, Juweid ME. Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25:579–586.
    1. Dohner H, Estey EH, Amadori S. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–474.
    1. Wilson WH, Young RM, Schmitz R. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922–926.
    1. Le Tourneau C, Kamal M, Tsimberidou AM. Treatment algorithms based on tumor molecular profiling: the essence of precision medicine trials. J Natl Cancer Inst. 2016;108:djv362.
    1. Estey EH. Acute myeloid leukemia: 2012 update on diagnosis, risk stratification, and management. Am J Hematol. 2012;87:89–99.

Source: PubMed

3
Abonnere