Review a list of Rapid Response Pilot Project awards and Functional Genomics Initiative Research Awards that have been funded by the Functional Genomics Initiative since it launched.

Rapid Response Pilot Project Awards

Rapid Response Pilot Project Awards provide seed money for investigators to quickly pursue promising research leads using new data emerging from the CMO. Applications for these awards, typically $25,000 each, are accepted by the Functional Genomics Initiative Executive Committee on an ongoing basis. Support to successful applicants begins within three to four weeks of submission. This unique mechanism allows Memorial Sloan Kettering investigators to act quickly on new leads and maximize the opportunity for advancement in a competitive time frame.

2022

Zachary Epstein-Peterson
Longitudinal analysis of cell-free DNA (cfDNA) in patient-derived tumor xenograft (PDTX) models of IDH2-mutated angioimmunoblastic T-cell lymphoma (AITL)

Minkui Luo
Functional Annotation of PPARg Methylation Revealed by Cancer Hotspot Arginine

Philipp Niethammer
Testing the role of oxoeicosanoid signaling in tumorigenesis in zebrafish

2021

Danwei Huangfu (Co-PI: Dinshaw Patel)
Structure-function analysis of QSER1, a newly identified regulator of DNA methylation

Dirk Remus
Structural and Functional Characterization of the checkpoint clamp- clamp loader complex

Omar Abdel-Wahab
Understanding the functional implication of cancer-associated mutations in snRNAs (small nuclear RNAs)

Andrea Ventura
Investigating the role extrachromosomal circular DNAs in cancer

2020

Giuseppe Giaccone
Using Zebrafish to Investigate the Function of GTF2IL404H Mutation in Thymic Epithelial Tumors

Scott Keeney
Functional analysis of Topoisomerase II mutations associated with human cancer

Christine Mayr
Regulation of PI3K heterodimer formation

Justin Perry
Co-opting lung cancer-prevalent KEAP1 BTB/POZ domain mutations to investigate CUL3 substrate specificity

Marilyn Resh
Regulation of Diacylglycerol acyltransferase (DGAT1) substrate specificity and activity

Thomas Vierbuchen
Identifying Key Functions of ARID1B in neuron-specific BAF complexes

Morgan Huse
RAC1P29S and Biophysical Vulnerability in Melanoma

Samie Jaffrey
Using Cancer Genome Data to Reveal Regulators of the m6A Epitranscriptome

Xiaojing Ma
Functional investigation of a hotspot mutation in UBR5-driven cancers

Hans-Guido Wendel
Exploring the role of the H3K4 methyltransferase SETD1B in lymphomagenesis and targeted therapies

2019

Rohit Chandwani
A Genetic Model for Ampullary Cancer Subtypes

Lorenzo Galluzzi 
Impact of apoptotic proficiency on immune tumor infiltration and patient survival

William Jarnagin (Co-PI James Harding)
Cell-free DNA as a Predictive Biomarker in Cholangiocarcinoma

Minkui Luo
Characterization of Methylarginine-antagonistic Cancer Mutations of FOXO1

Simon Mantha
Assessing the Influence of the Coagulome on Cancer Survival

Nikolaus Schultz
Identification and characterization of novel mutations in non-unique regions of cancer genes

Maria Jasin 
RAD51D and its role in homologous recombination and replication stress suppression: effect of MSK-IMPACT mutations

Maurizio Scaltriti
Role of ARID1A in mammary epithelial development

Neal Rosen
Functional characterization of nonkinase domain BRAF mutations identified by MSK-IMPACT

2018

Ping Chi
Understanding the functional impact of cohesin mutations in cancer

Bishoy Faltas (Weill Cornell Medicine)
Dissecting the role of co-occurring FGFR and cell cycle genomic alterations in the sensitivity of urothelial carcinoma to combined FGFR- and CDK4/6-inhibition

Yariv Houvras (Weill Cornell Medicine)
Leveraging cancer genetics to identify the mechanisms of adaptive resistance in RET rearranged cancer

Christopher Klebanoff
Genomic characterization of a “public” neoantigen derived from a PIK3CA hot spot mutation in primary and metastatic cancers

Marc Ladanyi
Amplification of SRC family kinase genes: potentially novel and targetable primary drivers of tumorigenesis

Minkui Luo
Quantitative modeling of the vulnerability of MTAP-deleted cancer cells upon PRMT5 inhibition

Charles Sawyers
Mechanisms of oncogenicity caused by loss-of-function mutations in the DIS3 RNA exosome subunit

Etay Ziv
Mechanisms of functional synonymous DAXX mutations

2017

Minkui Luo
Characterization of Cancer-associated CARM1 Variants

Marilyn Resh
Role of Porcupine (Porcn) acyltransferase amplification and mutation in Wnt-driven cancer

Mark Ptashne
Functional analysis of SMARCD1 hotspot mutations associated with human cancer

2016

Dirk Remus
Functional Characterization of non-catalytic mutations in DNA polymerase δ associated with cancer

Kathryn Anderson
DNA Polymerase δ in development and tumorigenesis

Sarat Chandarlapaty
The Role of SF3B1 Mutations in Breast Cancer and Chronic Lymphocytic Leukemia Pathogenesis

Minkui Luo
Characterization of Potential Oncogenic SET8 Mutants Implicated in Breast Cance

Song-Hai Shi
Function of PDGFRβ in the Developing Mouse Neocortex: Implications on Neocortical Expansion and Glioma

M. Bryan Tsou
A novel mitotic wait signal mediated by the oncoprotein TRIM37

2015

Omar Abdel-Wahab
Investigating the oncogenic potential of amplifications and mutations in XPO1

John Chodera
Probing the biophysics of rar RAS mutations

Morgan Huse
Tuning polarized cell migration through the Ras-PI3K axis

Julia Kaltschmidt
Interaction mapping of Ig domain adhesion molecules Contactin-5/Caspr4

Yueming Li
Role of IFITM3 in regulation of gamma-secretase and Notch signaling

Minkui Luo
Characterization of SETD2’s catalytic domain mutations implicated in kidney renal clear cell carcinoma

Marilyn Resh
Role of Hedgehog Acyltransferase (Hhat) amplification and mutation in cancer

Andreas Ventura
Investigating the role of UPF1 mutations in pancreatic adenosquamous carcinoma

John Chodera
Automated biophysical characterization of clinically observed kinase point mutations on inhibitor affinity

Mary Goll
Identifying targets of Tet2/Tet3 regulation in normal development and myeloid malignancy

Jerard Hurwitz
Functional analyses of tumor-derived missense mutations in human DNA polymerase epsilon

Scott Keeney
Functional analysis of MLH1 mutations associated with human cancer

Bryan Tsou
Regulation of sensory capability of primary cilia by centriole sub-distal appendages

Hans-Guido Wendel
Characterizing the consequences of TNFRSF14/HVEM receptor mutations in follicular lymphoma

Xiaolan Zhao
Delineating genome stability effects of cancer mutations at a unique domain of POLE

2014

Jaclyn Hechtman
Functional characterization of SOX9 alterations in colorectal carcinoma

Prasad Jallepalli
Mechanisms of Chk1-dependent stabilization of replication forks and genome protection

Maria Jasin
Functional analysis of DMP MSK-IMPACT RAD51 cancer mutations

Marc Ladanyi
Defining MDM2 as a therapeutic target in RET-rearranged lung adenocarcinoma with acquired resistance to the RET inhibitor cabozantinib

Eric Lai
Functional analysis of diverse Dicer1 hotspot mutations associated with human cancer

Ming Li
Molecular understanding of tumor-associated sestrin mutations

Christine Mayr
Regulation of surface protein expression through mutations affecting UDPL

John Poirier
Functional impact of the Rlf-Mycl1 gene fusion in a mouse model of small cell lung cancer using CRISPR/Cas9 genome engineering

Jorge Reis-Filho
Defining the mechanistic basis of the PRKD1 E710D mutation as a driver of genetic alteration

Functional Genomics Initiative Research Awards

Proposals for support of up to $200,000 per year for two to three years are considered. Successful proposals should address functional and mechanistic implications of genetic information flowing from the CMO. The scope of the proposals should include, but not be limited to, functional analyses of genomic data using model organisms, molecular or biochemical approaches, and bioinformatic analyses. Full Project Grants do not require that the research project received prior Pilot Project funding.

Year 8 - 2022

Role of Pax5 in maintaining B cell identity and repressing transdifferentiation

Jayanta Chaudhuri
Immunology Program

Role of lineage-specific transcription factor SOX10 in ferroptosis and melanoma

Xuejun Jiang
Cell Biology Program

Targeting alternative lengthening of telomeres (ALT) in cancer

Agnel Sfeir
Molecular Biology Program

Year 7 - 2021

Assessing the foreignness of mutation associated neoantigens

Luis Diaz

The role of SWI/SNF, BLM and FANCM in DNA replication

Iestyn Whitehouse
Molecular Biology Program

Peripheral T Cell Lymphoma Driven by Aberrant Metabolic Signaling and Transcription

Ming Li
Immunology Program

 

 

Year 6 - 2020

Genomic and Transcriptomic Determinants of BRCA1 or BRCA2 associated cancers

Simon Powell
Molecular Biology Program

(Co-PI Jorge Reis-Filho, Human Oncology & Pathogenesis Program)

Homologous recombination deficiency and the impact of RAD51C mutation type on function and reversion

Maria Jasin
Developmental Biology Program

Functional analysis of microglial mutations associated with neuronal damage in Histiocytosis and Alzheimer disease

Frederic Geissmann
Immunology Program

Functions of the H3K36 methyltransferases NSD1 and NSD2 in gene regulation

Kristian Helin
Cell Biology Program

Year 5 - 2019

AID-induced MHC-II mutations

Jayanta Chaudhuri
Immunology Program

Structure, function and deregulation of SWI/SNF chromatin remodeling complexes in cancer

Alexandros Pertsinidis
Structural Biology Program

Dissecting the oncogenic role of del13q14 in Chronic Lymphocytic Leukemia

Andrea Ventura
Cancer Biology & Genetics Program

Cellular and epigenetic consequences of two mutations in the tumor gene KMT2D

Alexandra Joyner
Developmental Biology Program

Year 4 - 2018

Effects of cancer-associated SOX17 mutations on cell lineage specification and tissue morphogenesis

Anna-Katerina Hadjantonakis 
Developmental Biology Program

Description: SOX17, an evolutionarily conserved transcription factor involved in the development of embryos, has also been identified as a tumor suppressor. It is mutated in cancers of internal organs, including endometrial cancer, colon cancer, esophageal cancer, gastric cancer, hepatocellular carcinoma, and lung cancer. Using a combination of sophisticated genetic manipulations and state-of-the-art single-cell genomics approaches, combined with functional studies in vivo in embryos and in organoids, we will investigate the function of tumor-associated mutations in SOX17.

Precise modeling of cancer-associated single nucleotide variants using CRISPR base editing

Scott Lowe
Cancer Biology and Genetics Program

Description: Cancer arises as a consequence of mutations in genes that promote cell proliferation, survival, and metastasis. DNA-sequencing efforts have cataloged the mutations that can occur in individual human cancers, and indeed, most people treated at Memorial Sloan Kettering are now tested for the gene mutations that are present in their tumors. These cataloging efforts have identified hundreds of genes that can be altered in human tumors, often through different types of mutations in the same gene and in a range of different gene combinations. These efforts have helped better classify tumors and, in some cases, suggested therapeutic strategies that can be tailored to individuals with cancer. Still, to achieve the full benefit of this information, it is important to understand the functional consequence of specific mutations, such as how each alteration alters normal cell physiology and how mutant gene combinations produce distinct cancer behaviors. Our project aims to develop a suite of tools that will facilitate engineering the range of cancer-associated mutations observed in MSK’s patients for investigators to study. Eventually, we plan to develop strategies to target these mutations. This approach takes advantage of new genome-editing technology using CRISPR that has been optimized by the team to efficiently produce cancer-associated mutations in cultured cells and animal models. To illustrate the potential use of these tools, we will generate and study a series of mutations in the p53 tumor suppressor, which is the most frequently mutated gene observed in human tumors. These efforts will produce tools that will enable functional cancer research throughout the MSK community and will yield biological insights into a fundamentally important cancer gene. 

Deciphering the role of ADAM proteases in Notch and EGFR pathways using the genetic data from the CMO and TCGA

Dimitar Nikolov 
Structural Biology Program

Description: ADAM metalloproteases regulate normal and tumorigenic signaling by cleaving cell-surface substrates, including Notch, EGFR, and their ligands. Our proposed studies are aimed at understanding the molecular mechanisms of ADAM protease regulation and specifically the role of conformational ADAM ectodomain rearrangements in substrate recognition and cleavage. We will utilize the large amount of available CMO sequencing data from patients’ tumors with mutations in the substrate-binding domains of ADAM10 and ADAM17. We will study how ADAMs switch between “opened” and “closed” conformations and how this affects the downstream Notch and EGFR signaling.

The roles of BLM and FANCM homologs in attenuating replication stress

Dirk Remus
Molecular Biology Program

(Co-PI: Xiaolan Zhao)

Description: Mutations in the human FANCM and BLM DNA helicases underlie severe cancer predisposition syndromes. Consistent with their role in protecting the genome from cancer-causing mutations, previous studies have demonstrated an evolutionarily conserved role for FANCM and BLM helicases in the repair of DNA lesions. Here, exploiting the genetic and biochemical tractability of their budding yeast orthologues, we will investigate a recently emerged novel role for these helicases in promoting DNA replication during replication stress. This condition is often observed in cancer cells and is characterized by the impairment of replication forks. These studies will enhance our understanding of the mechanisms that link genome instability and cancer.

Immunomodulatory function of E-cadherin in cancer

Ming Li
Immunology Program

Description: With immunotherapy being actively adopted for cancer care, a better understanding of tumor-elicited immune responses and tumor immune escape mechanisms is crucial for the development of new cancer treatments and for stratifying people for effective therapy. Our recent studies have revealed a novel tumor-resident cytotoxic lymphocyte response that’s broadly induced in human and murine malignancies. The current proposal will investigate how tumor-expressed E-cadherin supports the cancer immunosurveillance response and whether the E-cadherin mutations seen in patients’ tumors promote tumor immune escape.

Integrated approaches annotate functions of cancer-associated H3K36 methyltransferases

Minkui Luo 
Chemical Biology Program

(Co-PI: John Chodera)

Description: Among frequent cancer mutations on the MSK-IMPACT™ list are those associated with H3K36 protein methyltransferases (PMTs). This project will implement the integrated computational-experimental approach to elucidate the molecular mechanisms through which point mutations of H3K36 PMTs impact downstream functions. Our work will be essential to annotate gain/loss-of-function variants for PMTs.

Year 3 - 2017

Defining the oncogenic impact of ATP6AP1- and ATP6AP2-inactivating somatic mutations in granular cell tumors

Jorge Reis-Filho
Pathology Department

(Co-PIs: Britta Weigelt and Fresia Pareja)

Description: Granular cell tumors are rare lesions composed of cells that contain numerous granules whose nature is unknown. We have sequenced these tumors and found that the vast majority of them have mutations in the genes ATP6AP1 or ATP6AP2, which render them inactive. These genes, albeit not previously linked to cancer, play pivotal roles in the trafficking of materials inside cells. Our preliminary analyses have shown that inactivation of ATP6AP1 or ATP6AP2 in normal cells grown in petri dishes results in the formation of granules similar to those found in granular cell tumors. In this project, we will be seeking to develop models to study granular cell tumors and to understand how mutations in ATP6AP1 and ATP6AP2 result in the development of these tumors and in the production of the characteristic granules.

The determinants of immunogenicity of mutationally derived neoantigens of human cancers

David Scheinberg 
Molecular Pharmacology Program

(Co-PI: Andrea Schietinger)

Description: There are hundreds to thousands of genetic mutations detected in cancers, yet few of them can be treated with existing drugs. In spite of this, tumors with the highest number of mutations respond best to immunotherapies, even though we generally do not know which of the mutations are the target of the immune system. In this proposal, we seek to discover the rules for predicting which tumor mutations are immunogenic. If successful, we can apply these rules to the mutations found by the Marie-Josée and Henry R. Kravis Center for Molecular Oncology (CMO). We have developed a high-throughput, pooled-screening method (called PresentER) to discover which major histocompatibility complex (MHC)-presented protein sequences can be effectively recognized by human killer T cells and possibly cause tumor rejection. Longer term, this method may allow the CMO to vastly increase the number of therapeutically actionable mutations that are discovered, as well as to design new immunotherapies based on vaccines, engineered T cells, and T cell receptor mimic antibodies.

Functional requirement for DSS1 and the DBD domain of BRCA2 in maintenance of genome integrity

Maria Jasin
Developmental Biology Program

Description: The tumor suppressor gene BRCA2 is known to be mutated in families with a history of breast and ovarian cancer. However, more recently BRCA2 has been found to be mutated in prostate and other tumors in individuals with no family history of the mutations. This grant focuses on understanding the role of the DNA-binding domain of the large BRCA2 protein to clarify its role in tumor suppression, as well as its sensitivity to therapy and acquired resistance.

Understanding and exploiting replication stress in cohesin-deficient cancers

Prasad Jallepalli
Molecular Biology Program

Description: Our project investigates the impact of patient-derived mutations in cohesin, a ring-shaped complex that traps and tethers pairs of DNA molecules. While most famously involved in mitotic cell division, cohesin is now appreciated to have a direct role in DNA replication itself, mainly through work from our laboratory. In this project, we take advantage of our new discovery that two different cohesin subunits (STAG2 and PDS5B) control the speed and stability of replication forks (sites of DNA synthesis) and also affect cellular sensitivity to drugs that induce replication stress, a state in which replication fork integrity is partially compromised. This situation is commonly seen in cancer. Our studies will illuminate the link between cancer-derived cohesin mutations and replication stress, possibly suggesting routes for selectively targeting such cancers therapeutically.

The role of the oncoprotein TRIM37 in the surveillance of S phase and mitosis

Meng-Fu Bryan Tsou
Cell Biology Program

Description: Our preliminary studies found that two genes involved in promoting cell fitness are frequently amplified in human tumors, as indicated in the Center for Molecular Oncology’s data. This novel surveillance pathway is likely essential for the long-term fitness and robustness of cancer cells. Our proposal aims to understand the molecular mechanism by which cells maintain their efficiency and fitness in both normal and disease conditions.

Year 2 - 2016

ATM as a molecular rheostat during class switch recombination

Jayanta Chaudhuri
Immunology Program

Description: DNA breaks, especially DNA double strand breaks, are one of the most toxic lesions that can occur in a cell. Double strand breaks occur in a cell following genotoxic stress such as exposure to ionizing radiation or as mistakes during replication. If unrepaired, double strand breaks either lead to cell death or participate in aberrant DNA transactions leading to cancer. Despite the toxicity, for development of a robust immune system, B cells go through a process called class switch recombination in which DNA double strand breaks are deliberately introduced into the genome. A failure to generate such breaks lead to immunodeficiency syndromes while impaired repair leads to B cell lymphomas. Thus, mechanisms that regulate class switch recombination is highly relevant to both immunity and preservation of genomic integrity. We have discovered that a DNA repair protein, ATM, participates in both the generation and the repair of DNA breaks during class switching. However, the molecular mechanism by which ATM executes this dual function is completely unknown. This proposal uses data available from the MSKCC Functional Genomic database to elucidate the role of ATM in promoting immunity through the precise and controlled generation of DNA breaks and maintaining genomic integrity during this otherwise dangerous process. Successful completion of the project will illuminate novel pathways that will have major impact in our understanding of the ontogeny and intervention of B cell lymphomagenesis. 

Quantifying and Correlating Tissue Pathology with the MSK-IMPACT Genotype

Thomas Fuchs
Department of Pathology

(Co-PI Hikmat Al-Ahmadie, Nikolaus Schultz, Sahussapont Joseph Sirintrapun)

Description: The histological slides of samples that have undergone MSK-IMPACT testing are a prime example of “dark data” — stored, but unleveraged. Tapping into this terabyte-scale resource will provide unparalleled possibilities for novel, quantitative research for MSK scientists and clinicians.  We will apply and develop machine learning algorithms based on ensemble classifiers and deep learning to statistically describe and quantify tumor morphology from image data. This allows us to automatically correlate the resulting phenotype descriptors with genes or alleles from MSK-IMPACT. With this framework we can not only automatically screen for correlations but answer questions like: “Can we predict a genotype from a specific tissue morphology?”

Dysregulation of the Nutrient-Sensing Pathway in Cancer

Ming Li
Immunology Program

Description: Uncontrolled cell growth is the most fundamental feature of cancer.  In this proposal, we will define whether and how tumor cells acquire growth advantage by mutating key molecules involved in nutrient sensing.  These studies can help to stratify cancer patients for treatment with drugs targeting the nutrient signaling pathway.  

Delineating the impact of 12-lipoxygenase deletion on epithelial maintenance, inflammation and transformation in live zebrafish

Philipp Niethammer
Cell Biology Program

Description: 12-lipoxygenase is a scarcely studied member of an enzyme cascade that produces powerful bioactive lipids involved in inflammation, epithelial barrier maintenance, and tumorigenesis. Cancer genome database analysis (cbioportal.org) shows that 12-lipoxygenase is absent in various tumors due to a chromosome deletion. Using genomic and live imaging approaches in zebrafish, we will test causal connections between 12-lipoxygenase deletion, loss of epithelial barrier integrity, epithelial inflammation, and tumorigenesis to identify potential tumor suppressive functions for this enzyme.

The mechanism of eukaryotic initiation factor 4E (eIF4E) mediated translational control

Xiaohui Qu
Molecular Biology Program

Description: Eukaryotic initiation factor 4E (eIF4E) overexpression is oncogenic and it is commonly identified in cancers including several studies in CBioPortal. Interestingly, despite its characterization as a general translation initiation factor, eIF4E overexpression was found to selectively stimulate the translation of a subset of mRNAs that are essential for tumorigenesis. In this proposal, we will use a novel approach that combines our newly developed single-molecule translation assay with genomic RNA sequencing to better understand the molecular mechanism of eIF4E-mediated translational control of gene expression.

Understanding and Targeting Mutations in MEK1/2 in Cancer

Omar Abdel-Wahab
Human Oncology and Pathogenesis Program

(Co-PI Neal Rosen)

Description: Genetic alterations which result in activation of the “Mitogen Activate Protein Kinase” (“MAPK”) pathway are amongst the most common alterations in cancer. Normally, the MAPK pathway communicates signals from receptors on the surface of a cell to the DNA in the nucleus of the cell. In cancer, however, this pathway is often altered in a manner which results in hyperactivation of this pathway. The MAPK is also sometimes referred to as the “Ras-Raf-MEK-ERK pathway” after the names of each of the proteins in the chain of proteins that communicate signals in this pathway. While activating mutations in the first 2 members of this chain (RAS and RAF) are very well studied in cancer, mutations affecting the 3rd member of this chain, MEK, are far less understood. This is in part due to the fact that, until very recently, MEK proteins were thought to be rarely mutated at a high frequency in any one common form of cancer. However, the increased understanding of the genetics of many forms of cancer have highlighted that MEK mutations are seen consistently at a low frequency across many forms of cancer and are also common in a few rare forms of cancer. Thus, MEK-mutant cancers appear to represent an important subtype of cancer in aggregate. Moreover, at least a proportion of MEK mutations appear to sensitize cancer patients to specific drugs which inhibit MEK. MEK mutations may therefore be extremely important to understand clinically. Through generous funding from the Functional Genomics Initiative of MSKCC, we now aim to systematically understand the consequences of MEK mutations across a wide variety of cancers. We are studying the effects of these mutations on activation of the MAP kinase pathway, biological effects in cancer formation models, and response to inhibitors of MEK. We believe this work may be very important in promoting clinical efforts to treat cancer patients with MEK inhibitors and help promote the use of more selective and efficacious therapies for cancer patients. 

Molecular genetic analysis of cancer hotspot mutations in core miRNA machinery

Eric Lai
Developmental Biology Program

(Co-PI Danwei Huangfu)

Description: microRNAs (miRNAs) are ~22 nucleotide (nt) RNAs that mediate broad networks of gene regulation. Notably, cancer genome sequencing efforts (such as the TCGA and MSKCC-IMPACT projects) reveals somatic hotspot mutations that affect core factors in the miRNA pathway. We will conduct an integrated set of biochemical, genetic, and computational studies to understand what effects these mutations have on miRNA production and cellular behavior. Our goal is to use this information to understand why these mutations in miRNA machinery are beneficial to particular tumors, as this could provide a molecular framework for treating patients with these cancer signatures.

Year 1 - 2015

Functional analysis of cancer-associated PPP2R1A mis-sense mutations during cell division

Emily Foley
Cell Biology Program

Description: While most human cells have 46 chromosomes, tumor cells often have more or fewer chromosomes, a condition known as aneuploidy. Tumor sequencing data indicate that mutations and deletions in one protein family, called protein phosphatase 2A, positively correlate with aneuploidy. We aim to define the molecular mechanisms linking these cancer-associated aberrations to aneuploidy, which may provide insight into tumor development and growth.

The functional characterization of RhoA mutations found in diffuse gastric cancer

Alan Hall
Cell Biology Program

Description: Diffuse gastric cancer is a highly malignant adenocarcinoma of the stomach wall. Recent genomic sequencing efforts have revealed a significant incidence of somatic mutations in RhoA (23 percent in one study). Our goal is to characterize the functional significance of RhoA mutations, using both epithelial cell lines as well as patient-derived gastric tumor cells.

Mechanisms of mTOR activating mutations in cancer

James Hsieh 
Human Oncology and Pathogenesis Program

(Co-PI John Chodera)

Description: mTOR inhibitors (mTORi) have been approved for the use to treat kidney cancer and ER+ breast cancer. However, sporadic cases of exceptional benefits drawn from mTORi for patients of other cancer types have been reported, indicating the need to select genomics-defined patients for precision medicine. We have discovered a series of mTOR activation mutations in kidney cancer patients and characterized their activities. Importantly, these mutations remain sensitive to mTORi. In this proposal, we mined the CMO dataset, which reports 388 mTOR missense mutations. There are 192 (~50 percent) mutations residing at the FAT or kinase domains. Importantly, most mutations either appear in clusters or focus at evolutionarily conserved positions. The remaining 180 are dispersed throughout the HEAT domain.

Hence, we propose to systemically interrogate these cancer-derived mTOR mutations. We hypothesize that a number of the mutations in FAT and kinase domains and certain mutations that were observed at evolutionary conversed positions in the HEAT domain are gain-of-function cancer driver mutations. With clear functional annotation of these mutants, oncologists could soon prescribe mTORi for tumors carrying activation mTOR mutations.

Mechanisms of genomic instability and epigenetic dysregulation in rare cancers with cohesion complex mutations

Prasad Jallepalli
Molecular Biology Program

Description: The ring-shaped cohesin complex plays vital roles in shaping, expressing, transmitting, and repairing the genome. As such, recent sequencing efforts at MSK and elsewhere have unearthed frequent alterations in cohesin subunits in brain, bladder, soft tissue, and hematologic malignancies. Nonetheless, the precise function(s) of cohesin that are disrupted by these mutations, and their impact on the response to cytotoxic chemotherapy, remain poorly understood. In this project we will use genome editing to model and correct cancer-associated cohesin mutations in cultured cells. Using these systems, we will explore pathogenetic mechanisms and test strategies for enhancing therapeutic potency.

Deciphering the molecular mechanisms of Eph signaling using the genetic data from the CMO

Dimitar Nikolov
Structural Biology Program

Description: Our studies take advantage of the availability of large amounts of CMO sequencing data from tumors to study the molecular mechanisms of Eph receptor signaling and to understand how its misregulation causes cancer. The focus of our studies are three receptors, EphA3, EphA5, and EphB1, which are often mutated in multiple myeloma, lung adenocarcinoma, and melanoma, and which will be investigated using a combination of genomic, cell-biological, biochemical, and structural approaches.

Understanding the role FOXA1 in prostate cancer growth, transdifferentiation, metastasis and drug resistance using a novel prostate organoid system

Charles Sawyers
Human Oncology and Pathogenesis Program

Description: Recent DNA sequencing studies of hundreds of prostate cancer samples obtained from men with localized or metastatic disease have identified recurrent mutations in a gene called FOXA1. FOXA1 collaborates with another important gene encoding the androgen receptor, which is the target of hormonal therapies in prostate cancer. We hypothesize that mutations in FOXA1 perturb the normal function of the androgen receptor, setting the stage for prostate cancer to develop and develop resistance to hormone therapy. We will test this hypothesis using a novel prostate organoid system that allows us to grow prostate cells obtained from patients in the lab. The results may also have implications in breast and lung cancer, where alterations in FOXA1 are also frequently observed.

Functional consequences of BLM cancer mutations to genome maintenance

Xiaolan Zhao
Molecular Biology Program

(Co-PI Maria Jasin)

Description: Mutations in the DNA helicase BLM underlie the pathology of Bloom syndrome, one of the most penetrant cancer predisposition syndromes demonstrating genome instability. A better understanding of the role of BLM could provide vital insight into the link between genome instability and tumorigenesis, as well as inform the development of potential diagnostic and treatment strategies. We propose to examine BLM cancer mutations at the highly conserved residues identified in the CMO genomics dataset in gastric and ovarian cancers, in both yeast and mammalian model systems, to elucidate molecular mechanisms of BLM for genome maintenance.