Abstract

PURPOSEMany cancers can be treated with targeted therapy. Almost inevitably, tumors develop resistance to targeted therapy, either from pre-existence or by evolving new genotypes and traits. Intratumor heterogeneity serves as a reservoir for resistance, which often occurs as a result of the selection of minor cellular subclones. On the level of gene expression, clonal heterogeneity can only be revealed using high-dimensional single-cell methods. We propose using a general diversity index (GDI) to quantify heterogeneity on multiple scales and relate it to disease evolution.MATERIALS AND METHODSWe focused on individual patient samples that were probed with single-cell RNA (scRNA) sequencing to describe heterogeneity. We developed a pipeline to analyze single-cell data via sample normalization, clustering, and mathematical interpretation using a generalized diversity measure, as well as to exemplify the utility of this platform using single-cell data.RESULTSWe focused on three sources of patient scRNA sequencing data: two healthy bone marrow (BM) donors, two patients with acute myeloid leukemia—each sampled before and after BM transplantation, four samples of presorted lineages—and six patients with lung carcinoma with multiregion sampling. While healthy/normal samples scored low in diversity overall, GDI further quantified the ways in which these samples differed. Whereas a widely used Shannon diversity index sometimes reveals fewer differences, GDI exhibits differences in the number of potential key drivers or clonal richness. Comparison of pre– and post–BM transplantation acute myeloid leukemia samples did not reveal differences in heterogeneity, although biological differences can exist.CONCLUSIONGDI can quantify cellular heterogeneity changes across a wide spectrum, even when standard measures, such as the Shannon index, do not. Our approach can be widely applied to quantify heterogeneity across samples and conditions.

Highlights

  • In many cancers, there still exists a critical need to understand the mechanisms of the evolution of therapy resistance

  • MATERIALS AND METHODS We focused on individual patient samples that were probed with single-cell RNA sequencing to describe heterogeneity

  • We focused on three sources of patient single-cell RNA (scRNA) sequencing data: two healthy bone marrow (BM) donors, two patients with acute myeloid leukemia—each sampled before and after BM transplantation, four samples of presorted lineages—and six patients with lung carcinoma with multiregion sampling

Read more

Summary

Introduction

There still exists a critical need to understand the mechanisms of the evolution of therapy resistance. Acute myeloid leukemia (AML) is an aggressive hematologic malignancy the hallmark of which is the proliferation of immature myeloid cells in the bone marrow and life-threatening ineffective hematopoiesis.[1] AML is the most common adult leukemia, with an incidence of approximately 20,000 cases per year and a 5-year survival of only 26%.2,3. Diagnosis of AML requires greater than 20% of myeloid immature cells (myeloblasts) in peripheral blood or bone marrow. Median survival of untreated AML is measured in weeks.[4] Several AML targeted therapies have been recently approved—for example, midostaurin for patients with FLT3 mutated disease and enasidenib for those with mutations in IDH2.5,6 These mutations occur at rates of 25% (FLT3) and 5%

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call