Past Projects & Shared Equipment

Share

The Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center is pleased to have funded the following MSK investigators and specific metastasis-related projects and resource acquisitions.

2020

Simon Schwoerer (GMTEC Postdoctoral Researcher Innovation Award)
Investigating metabolism in the tumor stroma reveals an unexpected role of protein catabolism in pancreatic cancer

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive tumor characterized by nutrient-deprivation in the tumor microenvironment. How PDAC cells continue to grow despite nutrient deprivation is not fully understood. In this project, we are going to investigate whether the of uptake and degradation of extracellular proteins in non-malignant stromal cells can support PDAC progression by proving an alternative source of free amino acids.

Francisco Sanchez-Rivera (GMTEC Postdoctoral Researcher Innovation Award)
Reconstructing complex cellular trajectories in vivo with single cell resolution

Reconstructing complex cellular trajectories in vivo with single cell resolution The overarching goal of this project was to develop and apply methods to 1) define the molecular trajectories that cells navigate in vivo during acquisition of their phenotypic identity, 2) investigate whether these trajectories exhibit significant spatiotemporal plasticity and dynamics, and 3) assess whether disease-associated genetic alterations (e.g. cancer mutations) influence these trajectories. While classical lineage tracing approaches have provided significant insight into these questions, achieving a complete and dynamic understanding of complex cellular trajectories and cell state transitions in vivo requires quantitative recording, measurement, and reconstruction of phylogenetic and transcriptional relationships over time with single cell resolution. Towards this goal, we developed an experimental framework in close collaboration with the group of Dana Pe’er that combines single cell RNA sequencing (scRNA-seq) and CRISPR-based genetic barcoding to dynamically characterize the emergence, plasticity, and lineage relationships of cancer cell populations in vivo. This system, which we refer to as Single-cell Transcriptomics And lineage Reconstruction (STAR), is based on a first-principles approach grounded on the hypothesis that each sgRNA has intrinsic editing properties largely determined by the sequence and difficult to predict without functional interrogation. This led us to develop and deploy a molecular recording ‘sensor’ platform (adapted from base editing sensor strategies partially supported by GMTEC; see Sánchez-Rivera et al., Nature Biotechnology, 2022) to identify sgRNAs with exceptionally high allele diversity and low large deletion probabilities that record at diverse editing rates. These optimized sgRNAs and cognate target sites are currently serving as a backbone to build an optimal lineage tracing system capable of achieving very high resolution and dynamic recording rates for robust lineage tracing over multiple temporal scales.

2019

Ming Li (Classic Individual)
Reprogram Tumor-Associated Macrophages to Heal the Tumor Wound

Tumors develop by invoking a supportive microenvironment resembling a non-healing wound characterized by aberrant angiogenesis and chronic infiltration of tumor-associated macrophages (TAMs). Here we show that TAMs can be reprogrammed to heal the tumor wound following ablation of TSC1, a repressor of the metabolic regulator mTORC1. We found that the tumor parenchyma-localized TAMs exhibited low mTORC1 activity. The depletion of TSC1 in TAMs led to mTORC1-mediated tumor suppression independent of the adaptive immune system. Instead, TSC1-deficient TAMs redistributed to a perivascular niche and rectified the tortuous and hyperpermeable vasculature, causing tumor tissue hypoxia and cancer cell death in distant avascular regions. Notably, TSC1-deficient TAMs exhibited reduced expression of inflammatory and angiogenic genes but increased expression of transcripts that promote macrophage metabolism and scavenger function. Thus, TAMs can be metabolically reprogrammed via TSC-mTORC1 signaling to an anti-inflammatory and pro-resolving state to reconfigure the tumor vasculature and suppress cancer progression.

2018

Minkui Luo (Classic Individual)
CARM1 Inhibition Alters the Epigenetic Plasticity of Metastatic Breast Cancer Cells

Numerous epigenetic events need to be orchestrated for breast cancer cells to metastasize to remote organs. Significant efforts have been made to identify the epigenetic cues that module metastatic outgrowth. CARM1 is an important epigenetic modulator to which metastatic breast cancer cells are addicted. High levels of CARM1 strongly correlate with breast cancer malignancy. We recently developed CARM1 chemical probes to interrogate CARM1-associated biology. The GMETC grant allowed us to evaluate in vivo efficacy of these compounds against breast cancer metastasis. While these treatments had no effect on the primary lung lesion, they significantly suppressed the secondary metastatic lesion and prolong overall survival. In addition, we conducted multiple sets of scRNA-seq analysis on gene expression profiles of MDA-MB-231 cells and develop in-house machine-learning algorithms dissect subpopulation-associated transcriptomic signatures of invasive cells. We concluded that the invasion capability of MDA-MB-231 cells mainly arises from an invasion-prone subset, 80% of which can be depleted by our CARM1 inhibitors. Differential expression analysis of scRNA-seq data further revealed the single-cell transcriptional signatures of metastasis-addicted genes such as MORC4, S100A2, RPL39, IFI27, ARF6, CHD11, SDPR and KRT18.

Scott Lowe (Classic Individual)
The epigenetic control of tumor ecosystem dynamics during PDAC progression

Pancreatic ductal adenocarcinoma (PDAC) is an invariably lethal cancer that arises in the context of a unique tumor microenvironment. This project aimed to examine the dynamic nature and regulation of intratumoral heterogeneity in pancreatic cancer, from its inception through metastasis. To do so, we implemented an integrative in vivo approach that combines single-cell technologies, state-of-the art computational analyses and unique mouse models of pancreatic cancer enabling perturbation of gene function at different disease stages. Our results uncovered that Kras gene mutation and tissue damage (pancreatitis) cooperatively remodel the chromatin landscape of the pancreatic epithelium to produce altered cellular states that distinguish the neoplastic process from physiological regenerative responses (Alonso-Curbelo et al., Nature 2021). Ongoing studies are incorporating novel computational methods that integrate transcriptional and chromatin information to predict cancer-initiating cells and their paths to overcoming key barriers to malignancy: initiation, benign to malignant transition, and metastatic dissemination. Collectively, our work indicates that large-scale epigenomic remodeling events integrating inputs from tissue context (inflammation) and genetics (driver gene mutations) direct the co-reprogramming of pancreatic epithelial cells and their niche early during tumor development, and establishes key cell- and non-cell-autonomous nodes that may pave the way for novel means to intercept and potentially revert the aberrant cell states responsible for PDAC initiation and metastatic progression.

2017

Paul Chapman with Kayvan R. Keshari, Taha Merghoub, Craig B. Thompson, Santosha Vardhana, Richard White, and Jedd Wolchok (Multi-Project)

Metabolic crosstalk between melanoma and its microenvironment

There have been astounding advances in the treatment of metastatic melanoma with both targeted therapy and immunotherapy, but unfortunately more than half of people with metastatic melanoma still die of the disease. Therefore, it is important to understand the mechanisms of melanoma metastasis. This wide-ranging effort will explore the role of cellular stress and immune responses on metastasis in melanoma. Researchers also will investigate how the melanoma microenvironment induces metabolic alterations within melanoma cells that directly facilitate cellular invasion and metastasis.

This is the first GMTEC award to include multiple projects. In addition, the research team has applied to the National Institutes of Health for a P01 grant related to this work.

Project 1: Molecular mechanisms, immune responses, and cancer cell metabolism

Santosha Vardhana and Craig Thompson

This project will explore the metabolic consequences of redox regulation and its effect on T cell function within the tumor. It aims to look at the connection between metabolic adaptation in the setting of chronic inflammation and the promotion of immune evasion and metastasis in melanoma. The researchers will examine the role of altered metabolism in promoting immune dysfunction in melanoma. They also seek to determine the contribution of exogenous fatty acids to altered melanoma-specific immune responses.

Project 2: Characterize the role of lipid uptake–induced ER stress in metastasis of melanoma

Richard White

This project will examine the ability of melanoma cells to metabolize fat as an energy source despite BRAF inhibition. One tool in this research is transgenic zebrafish, which will be used to characterize the role of fat cells in the progression of melanoma. These models will also be used to assess the mechanism of lipid-mediated ER stress in melanoma invasion and study the effects that lipids have on immune cells within the tumor microenvironment. The project will also look at whether blocking SLC27A fatty acid transporter proteins has therapeutic potential, and may ultimately lead to clinical trials evaluating this approach.

Project 3: Enhancing immune-mediated control of melanoma metastasis by modulating glycolytic stress

Jedd Wolchok

This project will examine the LDH/lactate axis and the effect of lactate on immune suppression in melanoma. It aims to study the connection between lactate production and the anti-melanoma immune response. It will also investigate whether blocking lactate production can improve T cell responses against melanoma.

2016

Mary Baylies
Identifying novel drugs that inhibit aRMS metastasis.

Proliferation and Migration profiles of drug-treated aRMS tumor cells (RH30)

Figure: Proliferation and Migration profiles of drug-treated aRMS tumor cells (RH30). Panel A is a still image from a live-cell imaging analysis that automatically tracks individual cell shapes, velocities, cell divisions, and cell deaths. Lower panels show examples of proliferation (B) and migration (C) responses to different concentrations of single and combination drug treatments.

The most common pediatric soft tissue sarcoma is rhabdomyosarcoma (RMS), representing 3 to 5 percent of all childhood cancers. Alveolar RMS (aRMS) is its most aggressive form and is associated with expression of PAX3- or PAX7-FOXO1 fusion oncoproteins. Although successive clinical trials have improved survival rates for RMS patients, the outcome with standard treatment for those patients with metastatic or recurrent disease remains bleak. (The five-year survival rate is less than 20 percent.) Moreover, within the last 30 years, there have been no significant changes in treatment, and no precision treatments exist. Hence, there is a critical need to investigate RMS metastasis and to identify novel therapeutic agents for its treatment. This is the goal of the Baylies lab’s work.

With funding from MTECH, the Baylies lab has identified 17 FDA-approved or in-trial drugs that target aRMS metastasis. These drugs, combined with a novel, ongoing screen in a Drosophila aRMS model, reveal 11 pathways essential for disease development. The activity of eight of the identified drugs that affect the RAS-MAPK, PI3K-AKT, and NFkB signaling pathways has been confirmed in this in vivo model. Importantly, the Baylies lab has identified three combinations of these drugs that eliminate or significantly reduce the metastatic potential of aRMS.

Based on these and published data, the Baylies lab is testing the FDA-approved/in-trial drugs that target RAS-MAPK, PI3K-AKT, and NFkB for validation and further characterization in human RMS tumor cell lines and in an aRMS ‘metastasis’ xenograft model. For the human tumor cell lines, the Baylies lab has developed a high throughput, image-based approach in collaboration with Andrew Cohen at Drexel University to assess tumor cell behaviors upon drug treatment (single and combinatorial treatment). The Baylies lab has now identified several FDA-approved/in-trial drugs and drug combinations that preferentially affect different aspects of human metastatic tumor cell behavior, including adhesion, migration, proliferation, or survival. The Baylies lab is now examining these drug combinations in aRMS xenograft models that reproduce aspects of the metastatic process, including survival in the blood stream, extravasion, colonization of different tissue sites, and proliferation at these sites. These experiments provide the critical next step toward patient treatment. Successful completion of these experiments will provide new precision therapeutic agents and regimes for testing in the clinic.

2015

Scott Keeney
Topoisomerase II in tumorigenesis and cytotoxic chemotherapy.

Richard White
Development of microenvironmental CRISPRs for the study of metastasis.

2014

Johanna Joyce
Investigation of the Microenvironmental Transcriptome in Breast to Brain Metastasis.

Sarat Chandarlapaty
Role of ESR1 Mutation in Breast Cancer Metastases

Scott W. Lowe
Dissecting the interactions between Smad4 and p53 inactivation in pancreatic cancer metastasis

Kathryn Anderson
Strip1 and STRIPAK complexes in cell migration and metastasis

2013

Mary Baylies
Finding New Treatments for Rhabdomyosarcoma

Colin Begg
Contralateral Breast Cancers: Independent Cancers or Metastases?

Richard White
Metastasis in the Zebrafish System

2012

Robert Benezra
Neutrophil-Mediated Antimetastatic Therapy

Johanna Joyce
Developing Novel Strategies to Target Tumor Cell-Macrophage Interactions in Brain Metastasis 

Daniel A. Heller
Real-Time In Vivo Marker Detection for Rapid Measurement of Metastasis Onset

2011

Morgan Huse
Exploring the Anti-Tumor and Anti-Metastatic Function of Protein Kinase C-θ Microclusters in Natural Killer Cells

Alexander Rudensky
Regulatory T cells and Breast Cancer

David A. Scheinberg 
Does Ablation of Tumor Neo-Vasculature Promote or Inhibit Metastatic Potential?

2010

Timothy A. Chan
Large-Scale Analysis and Clinical Application of the Breast Cancer Metastasis Epigenome

Nai-Kong V. Cheung
Genetic Analyses of Secondary CNS Metastases in Neuroblastoma (NB)

Larry Norton
Identifying Clinical Tools to Detect SRC as a Critical Moderator of Bone Metastasis Latency in Women with ER Positive

Filippo Giancotti
Functional Genomics Analysis of Tumor Dormancy at Metastatic Sites


Shared Resources

  • The MSOT Acuity Echo for Clinical Optoacoustic Tomography
    Moritz F. Kircher, MD, PhD
  • An Image-Guided Microirradiator for Preclinical Radiation Research
    Joseph Deasy, PhD
  • The Ivis Spectrum for Pre-Clinical Oncology Research
    Pat Zanzonico, PhD