Abstract

3122 Background: Tumors are complex ecosystems where exogenous and endogenous cues are integrated to either stimulate or inhibit cancer cell proliferation. However, the nature of these complex cell cycle states, their spatial organization, response to perturbation, and implications for clinical outcomes, are poorly characterized in tumor tissues. Methods: We used multiplexed tissue imaging to develop a robust classifier of proliferation, the multivariate proliferation index (MPI), using 513 unique tumors across five cancer types. Next, we used dimensionality reduction analysis to assess how the patterns of cell cycle protein expression in tumors were altered in response to perturbation. Results: The MPI outperforms single markers, like Ki67, when classifying proliferative index across diverse tumor types and reveals the proliferative architecture of tumors in situ. We find that proliferative and non-proliferative cancer cells are organized across microscopic (cell-to-cell) and macroscopic (tissue-level) scales. Both domains are reshaped by therapy, and local clusters of proliferative and non-proliferative tumor cells preferentially neighbor distinct tumor-infiltrating immune cells. We further phenotyped non-proliferating cancer cells using markers of quiescent cancer cells, cancer stem cells, and dormant cancer cells. We found that these types of non-proliferating cancer cells can occupy distinct regions within the same primary tumor. In high-dimensional marker space, populations of proliferative cancer cells express canonical patterns of cell cycle protein markers, a property we refer to as “cell cycle coherence”. Untreated tumors exist in a continuum of coherence states, ranging from optimal coherence, akin to freely cycling cells in culture, to reduced coherence characterized by either cell cycle polarization or non-canonical marker expression. Coherence can be stereotypically altered by induction and abrogation of mitogen signaling in a HER2-driven model of breast cancer. Cell cycle coherence is modulated by neoadjuvant therapy in patients with localized breast cancer, and coherence is associated with disease-free survival after adjuvant therapy in patients with colorectal cancer, mesothelioma and glioblastoma. Conclusions: The MPI robustly defines proliferating and non-proliferating cells in tissues, with immediate implications for clinical practice and research. The coherence metrics capture the diversity of post-treatment cell cycle states directly in clinical samples, a fundamental step in advancing precision medicine. More broadly, replacing binary metrics with multivariate traits provides a quantitative framework to study temporal processes from fixed static images and to investigate the rich spatial biology of human cancers.

Highlights

  • Uncontrolled cell proliferation is a defining feature of cancer[1] much of our understanding of the cell cycle comes from the in vitro study of cell monocultures grown in abundant nutrients, supporting doubling times of 24-48 hours[2]

  • An Multivariate Proliferation Index (MPI) value was assigned to each tumor cell based on the following rule: cells were scored as proliferative (MPI +1) if they expressed a positive balance of proliferation markers; non-proliferative (MPI 0) if they lacked expression of proliferation markers; and arrested (MPI -1) if they expressed high levels of one or both of the arrest markers, even if proliferation markers were expressed

  • Ki-67 is the most widely used measure of proliferation in diagnostic and research settings[60,61], we found that 39-72% of MPI +1 cells were Ki-67 negative but positive for PCNA or MCM2, depending on the tumor type; this is consistent with data from cultured cells showing that high Ki-67 expression occurs in the G2 phase of the cell cycle[22,23] (Extended-Data-Fig.1b,c)

Read more

Summary

Results

A Multivariate Proliferation Index (MPI) derived from multiplexed immunofluorescence images. We performed CyCIF on a cohort of 26 breast cancer specimens (TMA1, TMA2), identified MPI +1 epithelial cells and used the ccD-CMD algorithm to quantify IOV and CFD for individual patients (Fig.4a,b). Proliferating breast cancers expressing the same oncogenic driver (HER2) can exhibit different cell cycle dynamics (Fig.4a) within a single specimen ranging from a canonical: IOVlow CFDlow state to skewed distributions consistent with cell cycle phase disruption (skewed: IOVhigh CFDlow) and states not normally encountered in normally growing cells (non-canonical: IOVlow CFDhigh). In specimens acquired from patients enrolled in a clinical trial (NCT02326974) of neoadjuvant dual HER2 therapy, only one sample had a detectable population of proliferating cells following treatment (Fig.5b) In this specimen, HER2 levels were lower than in any pre-treatment sample and the state was IOVhigh CFDlow (Fig.5c). We conclude that cycle coherence in mesothelioma and glioblastoma is associated with aggressive tumor behavior and worse progression free survival

Discussion
Data and code availability
Experimental methods
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.