Open access peer-reviewed chapter

Precision Medicine Concepts in T-Cell Lymphoma

Written By

Philipp Staber

Submitted: 03 May 2018 Reviewed: 28 February 2019 Published: 17 April 2019

DOI: 10.5772/intechopen.85543

From the Edited Volume

Peripheral T-cell Lymphomas

Edited by Pier Paolo Piccaluga

Chapter metrics overview

1,143 Chapter Downloads

View Full Metrics

Abstract

Modern oncology witnesses an increasing number of new effective anticancer drugs targeting specific oncogenic pathways. Despite these advances, real-world experience with targeted single agents is disappointing since drug resistance usually occurs after a short time. This is particularly true for patients with refractory or relapsed T-cell lymphoma (TCL) who so far could not benefit from novel agents and demonstrate a short survival time of only 3 months. The novel genetic information gained from genome-wide high-throughput techniques has greatly improved our understanding of TCL. However, if precision medicine strategies are based solely on genetics, it runs into two major challenges: (1) the heterogeneity within the cancer of an individual patient and (2) the incomplete understanding of the degree of contribution of a specific mutation to a tumor phenotype. Next-generation functional drug screening (ngFDS) aims to address these problems. Studies that proof the clinical utility of ngFDS are currently limited. The following chapter aims to discuss recent advances of ngFDS and to line out its potential for TCL patients.

Keywords

  • T-cell lymphoma
  • precision medicine
  • genomics
  • transcriptomics
  • next-generation functional screening

1. Introduction

TCL is a heterogeneous group of rare lymphoid malignancies generally with dismal outcome [1, 2]. With current treatment options, a majority of patients do not achieve remission or experience relapse after completion of therapy [3, 4, 5, 6]. Patients with newly diagnosed TCL are most commonly treated with anthracycline-based (CHOP-like) chemotherapy regimens, often followed by consolidation with high-dose chemotherapy and stem cell transplantation in eligible patients [1]. Patients with relapsed TCL have a dismal prognosis with a median overall survival of only 3 months [2]. Unfortunately, mechanisms of drug resistance in TCL leading to progression and relapse remain elusive, and predictive biomarkers do not exist, precluding clinical progress. Three major limitations have thus far hampered a systematic and causative understanding of drug resistance in TCL: (I) adherence to genetic studies and barriers in translating this genomic information into direct clinical benefit for patients, (II) as for functional analysis, a general adherence to cell lines that is prone to clonal artifacts and that not faithfully recapitulates relevant physiology, and (III) lack of considerable biobanks of viably frozen cells from fresh dissociated lymphomas. All three points are discussed in detail below.

Advertisement

2. Precision medicine concepts

2.1 Advances and shortcomings of genetic studies

Predicting clinical treatment outcome from detailed characterization of patient material is one of the key challenges of modern oncology. The most so-called precision medicine approaches either rely on correlation of clinical outcome with molecular profiles such as genetic mutations [7] or attempt to reproduce the disease in an ex vivo model system and extrapolate from measured drug response to the patient outcome [8, 9]. Genetic approaches are state of the art for most diseases and have also been successful in some indications (e.g., targeting BCR-ABL with imatinib in chronic myeloid leukemia (CML)). TCLs, however, as most cancers, categorically differ from the rare monogenetic disease model and are driven by microevolutionary processes leading to broad genetic heterogeneity [10] and making a purely correlational logic extremely challenging [11].

Recent sequencing studies in TCL confirmed a set of genetic alterations in specific subtypes, including recurrent mutations in the epigenetic modifiers TET2, IDH2, and DNMT3A and the small GTPase RhoA in angioimmunoblastic T-cell lymphoma (AITL) [12, 13, 14, 15], while JAK/STAT pathway alterations through mutations at various levels seem to be present across all TCL subtypes, particularly in anaplastic large cell lymphoma (ALCL) [16]. For the most common subtypes of TCL, including peripheral T-cell lymphoma not otherwise specified (PTCL-NOS), AITL, and ALCL, genetic data are largely based on targeted DNA sequencing approaches focusing on mutations in pre-selected panels of genes, while unbiased sequencing approaches like whole exome sequencing are only available in small cohorts. Previous studies in ALK- ALCL utilizing immunohistochemistry and fluorescence in situ hybridization identified for ~38% of the patients with ALK- ALCL DUSP22- and TP63-rearrangements to be associated with good prognosis as for DUSP22- or with exceptional dismal prognosis as for TP63- rearrangements when receiving CHOP-like regimens [17].

Despite the significant amount of recurrent mutations in genes involved in DNA methylation like DNMT3A, TET2, or IDH2, no genome-wide epigenetic profiling has been reported in a sufficient amount of clinically well-annotated samples [12, 13, 18]. Therefore, we currently only can assume that broad epigenetic changes are frequent in TCL. In contrast to B-cell lymphoma, relatively strong responses to epigenetic modifying agents, such as HDAC inhibitors and methylation modifiers, are clinically evident [19, 20, 21].

Thus, there is evidence that in TCL the epigenome is of particular importance; however, systematic studies on epigenetic profiling in TCL are lacking and so are direct connections of genetic mutations in epigenetic modifiers and clinical responses to epigenetic drugs. A comprehensive characterization of genetic and epigenetic alterations and clinical response to specific drugs is highly warranted for TCL.

2.2 Advances and shortcomings of functional studies

Despite significant success to identify genomic alterations that might establish and drive hematologic malignancies, genetic studies face problems in translating genomic information into drug-able targets to directly benefit the patients. Results of personal medical efforts in cancer patients so far are sometimes promising but with the majority rather appearing disappointing [22, 23, 24, 25, 26]. In a landmark study that used each patient as their own control, Von Hoff et al. reported that 27% of patients with recurrent metastatic cancer of any kind had a 30% longer progression-free survival (PFS) with treatment selected on the basis of genetic profiling than they did with their previous treatment [25].

The first randomized trial of genomic-based precision medicine, the SHIVA study, did not use patients as their own control but investigated the effect of genetic marker-based targeted treatment comparing with treatment at physician’s choice in heavily pretreated patients with cancer. The SHIVA study failed to demonstrate a benefit for patients treated with genetic precision treatment [26]. We need to consider that our knowledge of how cancer genotype relates to its phenotype and of the complexity of the dynamic microevolutionary procedures that occur in an individual cancer is very limited [10].

Therefore, dynamic approaches that measure drug responses in cancer cells derived from patient biopsies promise to complement the static molecular measurements. Functional approaches could contribute important information to improve the selection of the right drug for the right patient at the right time. For instance, ex vivo chemosensitivity tests have been performed in samples of patients suffering chronic or acute leukemia [8, 27, 28, 29, 30] and also in gut stem cell-derived organoids [31, 32], in breast cancer cell lines [33], and in xenografted mice [34, 35]. All these pioneering functional studies have not been integrated into clinical routine, but they provided proof of concept for ex vivo responses that may match clinical response. However, due to the extensive time of the assays used, the clinical benefits were limited [36, 37, 38]. First proof of concept data was obtained with functional screens detecting drug activity in cell cultures with luminescent assays that predicted activity and resistance to drugs in patients [9] and leading to initiation of a clinical trial for targeted therapy of relapsed acute myeloid leukemia [39].

In two recent reports, luminescent assay-based drug profiling has also been performed in a T-cell neoplasm and in T-cell prolymphocytic leukemia (T-PLL) [40, 41]. T-PLL is a rare, clinically heterogenous neoplasm, which is treated with alemtuzumab-based induction in patients with symptoms, followed by consolidative stem cell transplantation [42]. There are no randomized controlled trials that inform the management of T-PLL; thus it is an area of clear unmet need. Boidol et al. used single-cell suspensions from fresh bone marrow, peripheral blood, or lymph node samples from 86 patients and exposed cells to 106 different compounds to perform functional drug screening after 72 hours [41]. Cancer cell-specific responses were calculated from individual dose-response curves, and tissue microarrays were generated for comparative protein expression profiling (Figure 1).

Figure 1.

Workflow: After sampling, single cell suspensions are used for high-throughput drug-screening. Formaldehyde conserved samples are processed for tissue microarrays for comparative protein expression profiling.

BCL-2 inhibitor venetoclax exhibited the strongest differential response for T-PLL (Figure 2). Ex vivo responses to venetoclax significantly correlated with BCL-2 protein expression scores but not with scores for BCL-2 gene family members BCL-XL and MCL-1. BCL-XL and MCL-1 expression scores demonstrated a significant correlation, while only MCL-1 appears to be inversely correlated with BCL-2 expression. T-PLL samples were among the entities with the highest BCL-2 scores, showing the most dramatic response to BCL-2 inhibition via venetoclax. Importantly, the second ex vivo dug screening report on T-PLL also found consistent activities of BCL-2 inhibition in T-PLL, thus confirming these results in an independent cohort [40] (Figure 2). It also demonstrates the reliability and reproducibility of functional assays.

Figure 2.

Cancer cell-specific responses were calculated from individual dose-response curves in two independent publications. In both, drug-profiling revealed that BCL-2 inhibitor venetoclax exhibited a significant differential response for T-PLL.

It is noteworthy that luminescent assay-based functional assays can recapitulate differential responses to venetoclax of specific disease entities experienced in the clinic. High-resolution dose-response curves of venetoclax clearly distinguished CLL, AML, and T-PLL samples (Figure 3). As expected from clinical data, CLL samples demonstrated pronounced effects at already very discrete doses (50% inhibitory concentration [IC50], around 5 nM). In contrast AML samples responded at high concentrations (median IC50: 10 μM). Response curves of T-PLL samples were in the middle of AML and CLL samples (median IC50: 1 μM).

Figure 3.

High-resolution dose-response curves of venetoclax in CLL, AML, and T-PLL samples.

Trying to elucidate the mechanisms, we investigated the protein expression of other BCL-2 family members. Interestingly venetoclax treatment induced BCL-2 and BCL-XL protein expression in the two patients, since MCL-1 levels did not changed. Therefore BCL-XL upregulation could be a potential mechanism of venetoclax resistance, and the additional use of drugs targeting BCL-XL or suppressing BCL-XL could be mechanistically synergistic (Figure 4).

Figure 4.

Venetoclax treatment induced BCL-2 and BCL-XL protein expression.

Therefore, studies testing venetoclax with appropriate combination partners in T-PLL are warranted. Combination therapies are essential to overcome the resistance mechanisms that limit the long-term efficacy of conventional cytotoxic chemotherapies or targeted agents inhibiting single pathways. It is important to establish a workflow to systematically test multiple drug combinations and applied it to successfully identify synergistic combinations [43]. The main challenges to systematic large-scale drug-drug combination testing are that the number of two-drug combination remains in tens of thousands, thus limiting the numbers of combinations to be tested in primary patient material. To overcome this burden, the community continues to develop innovative computational approaches for preselecting sets of putative synergistic drugs involving network analysis, dynamic modeling, and high-content machine learning. However, based mainly on genetic data, only approx. 40% in silico predicted drug synergies are confirmed by ex vivo combination testing as demonstrated at the example of T-cell prolymphocytic leukemia (T-PLL) [44]. A reason for the rather low functional confirmation rate could be the low correlation between high-throughput ex vivo drug testing and mutation profiling [40]. It is therefore tempting to propose a smart way to preselect combination partners that are then analyzed in vitro.

2.3 Advances of single-cell functional studies

Luminescent assay-based functional assays have the big plus of high-throughput because full automation is more easily established. However, these assays cannot provide data at the single-cell level and thus no data on individual cell based functional information. Especially if the aim is to address the heterogeneity within one individual patient, i.e., to discriminate malignant and normal cells or to use complex co-culture systems, investigators could be interested in automated microscopic imaging technology. Minimally invasive protocols could provide drug-response information in co-culture systems. This could maintain leukemia and multiple myeloma cells for a longer cultivating time and enhance the screening capabilities of patient samples [45, 46, 47]. Snijder and colleagues investigated the clinical impact of a newly developed single-cell image analysis technology platform that operates using a combination of multi-parametric immunofluorescence and high-throughput automated microscopy [48]. In contrast to functional methods used previously, this next-generation functional drug screening (ngFDS) technique allows a fast tumor cell -specific quantification of biological parameters of millions of adherent and non-adherent individual cells with high sample efficiency, minimal sample manipulation, extensive automation, and fast turnaround times [49] (Figure 5).

Figure 5.

Next-generation functional drug screening (ngFDS) technique allows a fast tumor cell-specific quantification of biological parameters of millions of adherent and non-adherent individual cells.

The authors demonstrated that multi-parametric, image-based, immunophenotypic cytometry could reliably distinguish malignant cells from normal bystander cells in a high-content screening context. They showed how this approach can detect phenotypes across several cellular compartments, quantifying, for example, T-cell engagement by the bispecific, CD19-directed, T-cell engager blinatumomab in patient samples. They applied the method to patients with aggressive hematologic malignancies failing at least two lines of therapy and without further standard treatment options. These patients will usually receive either best available therapy or best supportive care or will be enrolled in clinical trials. Upon relapse, blood, bone marrow, pleural effusions, or excised lymph node biopsies were collected depending on the disease manifestation. The primary endpoints were to evaluate the feasibility of integrating ngFDS into the clinic and to assess clinical response in patients who received a treatment according to ngFDS results as an individual healing attempt [49] (Figure 6).

Figure 6.

Blood, bone marrow, pleural effusions, or excised lymph node biopsies were collected depending on the disease manifestation. The primary endpoints were to evaluate the feasibility of integrating ngFDS into the clinic and to assess clinical response.

This prospective single-center pilot study demonstrates that it is possible to integrate automated microscopy-based next-generation functional drug screening (ngFDS) for patients with aggressive hematologic malignancies into the clinical routine. Importantly, the ngFDS results suggested treatment regimens that lead to an improved ORR and longer PFS for patients than the last treatment regimen the patients had just experienced progression on (Figure 7). These results demonstrate that we are already in possession of a wide array of working chemotherapeutics and targeted inhibitors that in principle are capable of breaking drug resistance even in multi-refractory cancers, given that we identify the right drugs for each individual patient, at the right time during their treatment.

Figure 7.

ngFDS results suggested treatment regimens that lead to an improved ORR and longer PFS for patients than the last treatment regimen the patients had just experienced progression on.

The study name is EXALT for extended analysis for leukemia/lymphoma treatment. It is a prospective non-randomized study with each patient functioning as their own control, thus allowing to determine the overall effect across heterogeneous disease entities and different therapies. The nonexistence of randomization could harbor a bias. In future studies testing ngFDS-guided therapies, randomization and physician choice are warranted.

Image-based quantification of drug effects with single-cell resolution in patient biopsies, as introduced here, represents a robust and clinically useful platform to assign powerful individualized therapeutic regimens. The strength of the approach resides in the statistical power derived from monitoring with computer-aided precision millions of individual functional events, i.e., single-cell drug responses, combined with the ability to discriminate cell types, allowing to score specific rather than general and averaged cytotoxic effects. The approach may be valuable for the personalized identification of clinically effective therapies for many other hematological malignancies, especially for rare diseases, like TCL. The selection of personalized therapy by ngFDS benefits from the ability to measure hundreds to thousands of drug exposures using small patient samples, where each ex vivo treatment includes healthy-cell-controls from the same patient sample. This allows for direct estimation of the therapeutic window, while the minimal ex vivo culturing of cells and compatibility with clinical diagnostic markers ensure fast and relevant feedback. Further, the platform can take advantage of the use of liquid biopsies where small target cells can be detected. These integrated drug response profiles, or “chemotypes,” of individual people are the culmination of the interplay between a number of molecular parameters of the responding cells, including not only the genetic, proteomic, and metabolic state of the cells but also the direct and indirect molecular interactions with other cells [48], recapitulating relevant physiological complexity. Such chemotypes may offer functional insight into the underlying health status of an individual, with potentially wide-ranging implications also in preventative and participatory medicine.

As shown here, ngFDS can provide clinically useful guidance in the absence of genetic information. However, the full potential of the approach will certainly be realized only through the synergy with genomic and other molecular patient profiling. This could lead to highly accurate personalized treatments, coupled with companion diagnostics, as well as powerful route to mechanistic elucidation of gene-to-phenotype relationships.

2.4 Outlook for precision medicine in TCL

Collectively, the combination of advanced functional strategies, chemical genetics, and phenotypic screening approaches to holistically chart and chemically probe mechanisms of drug failure opens fundamental new treatment opportunities for TCL patients. Phenotypic single-cell drug screening is a highly innovative approach that helps to identify sensitive drugs and proved to be effective in late-stage hematological malignancies [49]. The next step to achieve the real treatment goal of a “long-term remission” is to identify a systematic way to identify mechanistically grounded effective drug-drug combinations. Clinically fully annotated viable sample sets consisting of sufficient TCL samples will provide the basis to probe with the help of network analysis, dynamic modeling, and high-content machine learning a useful algorithm for individual combination treatment to tackle drug resistance up-front.

Novelty emerges from the innovative intersection of cutting edge technologies, such as an innovative drug screening pipeline at cellular resolution, the use of novel immune competent xenograft mouse models that recapitulate human immune response, and single-cell RNAseq applied in primary TCL cells upon multiple drug perturbations. Foremost, it will be important to probe these data sets with cutting edge bioinformatics and network medicine pipelines.

ngFDS will give hemato-oncologists a new tool to identify the most promising treatment combinations to help overcome resistance in otherwise refractory TCLs on a patient-to-patient basis. The success of ngFDS, added a new layer to how doctors can base treatment decisions, since it significantly improved the patients’ outcome [49]. A rational guidance to pick the most appropriate combination partner promises to achieve long-term remissions.

I further expect that the established pipelines of ex vivo testing identified drug combinations will outline and instruct prospective clinical trials as has recently shown [41, 45] (approval and funding of a clinical study starting Q4 2018: venetoclax + ibrutinib in T-PLL (VIT-study;) Ph2 Study M18–803 (AbbVie) = the first global clinical trial for T-PLL).

Finally, ngFDS challenges the concepts of future clinical trial design. It requires a study design that provides the highest possible flexibility for experimental treatment to allow data-guided combination of targeted agents with antibody-based therapies for TCL. Larger prospective studies should focus on specific disease entities and randomize between arms of different treatment selection procedures to capture the full potential of functional assays for our TCL patients.

References

  1. 1. d'Amore F et al. Peripheral T-cell lymphomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2015;26(Suppl 5):v108-v115
  2. 2. Mak V et al. Survival of patients with peripheral T-cell lymphoma after first relapse or progression: Spectrum of disease and rare long-term survivors. Journal of Clinical Oncology. 2013;31(16):1970-1976
  3. 3. Swerdlow SH et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-2390
  4. 4. de Leval L et al. Angioimmunoblastic T-cell lymphoma is the most common T-cell lymphoma in two distinct French information data sets. Haematologica. 2015;100(9):e361-e364
  5. 5. Vose J et al. International peripheral T-cell and natural killer/T-cell lymphoma study: Pathology findings and clinical outcomes. Journal of Clinical Oncology. 2008;26(25):4124-4130
  6. 6. Moskowitz AJ, Lunning MA, Horwitz SM. How I treat the peripheral T-cell lymphomas. Blood. 2014;123(17):2636-2644
  7. 7. Schilsky RL. Personalized medicine in oncology: The future is now. Nature Reviews. Drug Discovery. 2010;9(5):363-366
  8. 8. Montero J et al. Drug-induced death signaling strategy rapidly predicts cancer response to chemotherapy. Cell. 2015;160(5):977-989
  9. 9. Pemovska T et al. Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia. Cancer Discovery. 2013;3(12):1416-1429
  10. 10. Friedman AA et al. Precision medicine for cancer with next-generation functional diagnostics. Nature Reviews. Cancer. 2015;15(12):747-756
  11. 11. Urbach D et al. Cancer heterogeneity: Origins and implications for genetic association studies. Trends in Genetics. 2012;28(11):538-543
  12. 12. Sakata-Yanagimoto M et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nature Genetics. 2014;46(2):171-175
  13. 13. Palomero T et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nature Genetics. 2014;46(2):166-170
  14. 14. Lemonnier F et al. Recurrent TET2 mutations in peripheral T-cell lymphomas correlate with TFH-like features and adverse clinical parameters. Blood. 2012;120(7):1466-1469
  15. 15. Cairns RA et al. IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma. Blood. 2012;119(8):1901-1903
  16. 16. Crescenzo R et al. Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. Cancer Cell. 2015;27(4):516-532
  17. 17. Parrilla Castellar ER et al. ALK-negative anaplastic large cell lymphoma is a genetically heterogeneous disease with widely disparate clinical outcomes. Blood. 2014;124(9):1473-1480
  18. 18. Wang C et al. IDH2R172 mutations define a unique subgroup of patients with angioimmunoblastic T-cell lymphoma. Blood. 2015;126(15):1741-1752
  19. 19. Amengual JE et al. A phase 1 study of romidepsin and pralatrexate reveals marked activity in relapsed and refractory T-cell lymphoma. Blood. 2018;131(4):397-407
  20. 20. Shustov A et al. Romidepsin is effective and well tolerated in older patients with peripheral T-cell lymphoma: Analysis of two phase II trials. Leukemia & Lymphoma. 2017;58(10):2335-2341
  21. 21. Cheminant M et al. Efficacy of 5-azacytidine in a TET2 mutated angioimmunoblastic T cell lymphoma. British Journal of Haematology. 2015;168(6):913-916
  22. 22. Khwaja A et al. Acute myeloid leukaemia. Nature Reviews. Disease Primers. 2016;2:16010
  23. 23. Hood L, Friend SH. Predictive, personalized, preventive, participatory (P4) cancer medicine. Nature Reviews. Clinical Oncology. 2011;8(3):184-187
  24. 24. Tsimberidou AM et al. Personalized medicine for patients with advanced cancer in the phase I program at MD Anderson: Validation and landmark analyses. Clinical Cancer Research. 2014;20(18):4827-4836
  25. 25. Von Hoff DD et al. Pilot study using molecular profiling of patients' tumors to find potential targets and select treatments for their refractory cancers. Journal of Clinical Oncology. 2010;28(33):4877-4883
  26. 26. Le Tourneau C et al. 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. The Lancet Oncology. 2015;16(13):1324-1334
  27. 27. Irish JM et al. Single cell profiling of potentiated phospho-protein networks in cancer cells. Cell. 2004;118(2):217-228
  28. 28. Kornblau SM et al. Dynamic single-cell network profiles in acute myelogenous leukemia are associated with patient response to standard induction therapy. Clinical Cancer Research. 2010;16(14):3721-3733
  29. 29. Lacayo NJ et al. 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. British Journal of Haematology. 2013;162(2):250-262
  30. 30. Frismantas V et al. Ex vivo drug response profiling detects recurrent sensitivity patterns in drug resistant ALL. Blood. Mar 16, 2017;129(11):e26-e37. DOI: 10.1182/blood-2016-09-738070. Epub 2017 Jan 25
  31. 31. Gao D et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176-187
  32. 32. van de Wetering M et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933-945
  33. 33. Yu M et al. Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science. 2014;345(6193):216-220
  34. 34. Hidalgo M et al. Patient-derived xenograft models: An emerging platform for translational cancer research. Cancer Discovery. 2014;4(9):998-1013
  35. 35. Roife D et al. Ex vivo testing of patient-derived Xenografts mirrors the clinical outcome of patients with pancreatic ductal adenocarcinoma. Clinical Cancer Research. 2016;22(24):6021-6030
  36. 36. Selby P, Buick RN, Tannock I. A critical appraisal of the "human tumor stem-cell assay". The New England Journal of Medicine. 1983;308(3):129-134
  37. 37. Von Hoff DD. Send this patient's tumor for culture and sensitivity. The New England Journal of Medicine. 1983;308(3):154-155
  38. 38. Samson DJ et al. Chemotherapy sensitivity and resistance assays: A systematic review. Journal of Clinical Oncology. 2004;22(17):3618-3630
  39. 39. Tyner JW et al. Kinase pathway dependence in primary human leukemias determined by rapid inhibitor screening. Cancer Research. 2013;73(1):285-296
  40. 40. Andersson EI et al. Discovery of novel drug sensitivities in T-PLL by high-throughput ex vivo drug testing and mutation profiling. Leukemia. 2018;32(3):774-787
  41. 41. Boidol B et al. First-in-human response of BCL-2 inhibitor venetoclax in T-cell prolymphocytic leukemia. Blood. 2017;130(23):2499-2503
  42. 42. Dearden C. Management of prolymphocytic leukemia. Hematology. American Society of Hematology. Education Program. 2015;2015:361-367
  43. 43. Licciardello MP et al. A combinatorial screen of the CLOUD uncovers a synergy targeting the androgen receptor. Nature Chemical Biology. 2017;13(7):771-778
  44. 44. He L et al. Patient-customized drug combination prediction and testing for T-cell Prolymphocytic Leukemia patients. Cancer Research. 2018;78(9):2407-2418
  45. 45. Fischer U et al. Genomics and drug profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia identifies recurrent mutation patterns and therapeutic options. Nature Genetics. 2015;47(9):1020-1029
  46. 46. Frismantas V et al. Ex vivo drug response profiling detects recurrent sensitivity patterns in drug-resistant acute lymphoblastic leukemia. Blood. 2017;129(11):e26-e37
  47. 47. Silva A et al. An ex vivo platform for the prediction of clinical response in multiple myeloma. Cancer Research. 2017;77(12):3336-3351
  48. 48. Vladimer GI et al. Global survey of the immunomodulatory potential of common drugs (accepted). Nature Chemical Biology. Jun 2017;13(6):681-690. DOI: 10.1038/nchembio.2360. Epub 2017 Apr 24
  49. 49. Snijder B et al. Image-based ex-vivo drug screening for patients with aggressive haematological malignancies: Interim results from a single-arm, open-label, pilot study. Lancet Haematol. 2017;4(12):e595-e606

Written By

Philipp Staber

Submitted: 03 May 2018 Reviewed: 28 February 2019 Published: 17 April 2019