Our Research

The DiFeo lab strives to make original and impactful contributions to the field of women’s cancer research.  Dr. DiFeo’s broad background in cancer genetics and biology and specific expertise in miRNA biology, chemotherapy resistance, tumor initiation, and development of patient-based models have positioned her laboratory to work on research projects that span the translational research continuum. Beginning with an in-depth analysis of patient tumors, then progressing to a functional assessment of key genetic drivers of ovarian cancer progression, and ultimately to the development of novel therapeutic approaches to abrogate these drivers to uncover therapies that will improve ovarian cancer patient survival. To accomplish this, we focus on three major research areas: 1) the generation of clinically relevant gynecological cancer models, 2) the discovery of potent genetic drivers with a focus on microRNAs involved in tumor initiation, drug resistance, and recurrence, and 3) development of novel or re-purposed drugs.


RESEARCH AREAS

Generation of Clinically Relevant Gynecological Cancer Models

Since the inception of the lab, we have devoted a substantial amount of time to fostering collaborations with clinicians to build a well-integrated research program that would promote patient-focused, impactful research ideas and permit the development of a large repository of gynecologic tumors and patient models. Through various gynecologic cancer tumor repositories across New York, Cleveland, and Michigan the DiFeo laboratory has been able to collect over 500 advanced-stage uterine and ovarian tumors. Most importantly, we have developed and fully characterized numerous patient-derived cell lines, organoids, and xenografts (PDX) which conserve original tumor characteristics such as heterogeneous histology, clinical biomolecular signature, malignant phenotypes, and genotypes. Given that several studies have shown that many of the commercially available ovarian cancer cell lines do not recapitulate the genomic signature of patient tumors this resource has been used by investigators across the country. We have shared these models with numerous collaborators in the scientific community as well as licensed these models to various CROs, which has led to the discovery of novel therapeutic targets, biomarkers, patent applications, and drugs.1-20 In addition, drug companies including Eli Lilly, Immunogen, SPARC/Sun Pharma, and Benevolent AI have collaborated on sponsored studies with the DiFeo laboratory further highlighting the distinctive feature of this resource.

 

Identifying Functional Drivers Involved in Ovarian Cancer Progression

The DiFeo played a central role in defining KLF6 and its novel oncogenic splice variant, KLF6-SV1, as a key regulator of several human malignancies.21-34 Our findings were the first to demonstrate that the large majority of ovarian cancer tumors have increased expression of the oncogenic KLF6-SV1 isoform and high levels of this protein correlated with increased tumor aggressiveness. Furthermore, through these studies, we discovered that KLF6-SV1 was a novel anti-apoptotic protein that targets the pro-survival molecule NOXA for degradation and its targeted inhibition extends survival ovarian cancer. These studies resulted in 18 publications during my doctoral and early post-doctoral studies in journals such as Science Translational Medicine, Journal of Clinical Investigation, Cancer Research, Clinical Cancer Research, and Oncogene. 21-34  

Currently, the DiFeo laboratory is focused on uncovering functional microRNA:mRNA pathways driving ovarian cancer chemotherapy resistance and disease recurrence. We uncovered that microRNA-181a is an oncoMIR wherein it is frequently overexpressed in recurrent, platinum-resistant high-grade serous ovarian cancer (HGSC) and correlates with shorter time to recurrence and poor overall survival. 35 Mechanistically, miR181a contributes to cellular transformation, metastasis initiation, and tumor recurrence by modulating chromosomal instability, EMT, and stemness factors via direct regulation of STING, TGFβ, and Wnt2-13. Most recently, we have shown that in both immunodeficient and immunocompetent HGSC mouse models, targeted inhibition miR181a resulted in decreased tumor dissemination, ascites accumulation, and increased sensitivity to immunotherapy. These studies have resulted in numerous publications including two in Nature Communications. In addition, this research provides the rationale for determining whether pharmacological inhibition of miR181a can simultaneously inhibit the key signaling pathways implicated in cancer progression and significantly enhance patient survival. In addition, several news outlets, including a blog published by the National Cancer Institute, have highlighted the research upon which this application is based. https://www.cancer.gov/news-events/cancer-currents-blog/2020/ovarian-cancer-form-microrna?cid=eb_govdel  Lastly, this work has resulted in the development of novel tools, such as a miR181a biosensor and a miR181a transgenic mouse, thereby laying the groundwork for future scientific endeavors and resources.

 

Development of Novel or Re-purposed Drugs to Treat Platinum-Resistant Cancer

The development of resistance to first-line chemotherapeutics most notably platinum-based therapies leaves few options for the clinical management of advanced ovarian cancer. Therefore, circumventing tumor resistance to commonly used first-line agents represents a very important aspect of multiple initiatives to eliminate ovarian cancer. Thus, another focus of the DiFeo laboratory is to uncover the underlying mechanism of platinum resistance and to develop therapeutic strategies to treat recurrent, chemotherapy-resistant diseases. Through the development of numerous platinum-resistant isogenic primary cell lines as well as patient-derived xenograft models we have uncovered several targetable pathways that can sensitize cells to platinum-based therapies.1-6,8,10,11,14,17-20, 36-39 For example, we are the first to show that altered glutamine metabolism contributes to platinum-resistant ovarian cancer and that targeting glutamine metabolism together with platinum-based chemotherapy offers a potential treatment strategy, particularly in drug-resistant ovarian cancer.4 Our lab has also used various drug repositioning platforms such as “DrugPredict” to identify FDA approved drugs for repositioning as ovarian cancer therapeutics. Using the DrugPredict algorithm, we have identified the Class III antiarrhythmic agent Amiodarone as a potential anti-cancer drug for ovarian cancer treatment6. These findings have allowed us to developed a novel anti-mitotic compound that decouples the potent calcium/potassium channel-blocking activities of this drug but retains the anti-tumorigenic effects.

Lastly, we have recently found that small molecule-mediated stabilization of protein phosphatase 2A (PP2A) modulates the Homologous Recombination pathway and potentiates DNA damage-induced cell death.20  These studies led to the discovery that PP2A modulators can surmount PARP inhibitor insensitivity, and that the combination of PP2A modulators and PARP inhibitors is a novel therapeutic approach for treating recurrent ovarian cancer.

REFERENCES

  1. Nagaraj AB, Joseph P, Kovalenko O, Singh S, Armstrong A, Redline R, Resnick K, Zanotti K, Waggoner S, DiFeo A: Critical role of Wnt/β-catenin signaling in driving epithelial ovarian cancer platinum Oncotarget.6(27): 23720-23734, 09/2015. PM26125441
  2. Wiechert A, Saygin C, Thiagarajan PS, Rao VS, Hale JS, Gupta N, Hitomi M, Nagaraj AB, DiFeo A, Lathia JD, Reizes O: Cisplatin induces stemness in ovarian cancer. Oncotarget.7(21): 30511-30522, 05/2016. PM27105520
  3. Hudson CD, Savadelis A, Nagaraj AB, Joseph P, Avril S, DiFeo A, Avril N: Altered glutamine metabolism in platinum resistant ovarian Oncotarget.7(27): 41637-41649, 07/2016. PM27191653
  4. Dimitrova N, Nagaraj AB, Razi A, Singh S, Kamalakaran S, Banerjee N, Joseph P, Mankovich A, Mittal P, DiFeo A, Varadan V: InFlo: a novel systems biology framework identifies cAMP-CREB1 axis as a key modulator of platinum resistance in ovarian cancer. Oncogene.36(17): 2472-2482, 04/2017. PM27819677
  5. Saygin C, Wiechert A, Rao VS, Alluri R, Connor E, Thiagarajan PS, Hale JS, Li Y, Chumakova A, Jarrar A, Parker Y, Lindner DJ, Nagaraj AB, Kim JJ, DiFeo A, Abdul-Karim FW, Michener C, Rose PG, DeBernardo R, Mahdi H, McCrae KR, Lin F, Lathia JD, Reizes O: CD55 regulates self-renewal and cisplatin resistance in endometrioid tumors. J Exp Med.214(9): 2715-2732, 09/2017. PM28838952
  6. Nagaraj AB, Wang QQ, Joseph P, Zheng C, Chen Y, Kovalenko O, Singh S, Armstrong A, Resnick K, Zanotti K, Waggoner S, Xu R, DiFeo A: Using a novel computational drug-repositioning approach (DrugPredict) to rapidly identify potent drug candidates for cancer treatment. Oncogene.37(3): 403-414, 01/2018. PM28967908
  7. Morrow JJ, Bayles I, Funnell AP W, Miller TE, Saiakhova A, Lizardo MM, Bartels CF, Kapteijn MY, Hung S, Mendoza A, Dhillon G, Chee DR, Myers JT, Allen F, Gambarotti M, Righi A, DiFeo A, Rubin BP, Huang AY, Meltzer PS, Helman LJ, Picci P, Versteeg HH, Stamatoyannopoulos JA, Khanna C, Scacheri PC: Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med.24(2): 176-185, 02/2018. PM29334376
  8. Belur Nagaraj A, Kovalenko O, Avelar R, Joseph P, Brown A, Surti A, Mantilla S, DiFeo A: Mitotic Exit Dysfunction through the Deregulation of APC/C Characterizes Cisplatin-Resistant State in Epithelial Ovarian Cancer. Clin Cancer Res.24(18): 4588-4601, 09/2018. PM29653924
  9. Noto FK, Adjan-Steffey V, Tong M, Ravichandran K, Zhang W, Arey A, McClain CB, Ostertag E, Mazhar S, Sangodkar J, DiFeo A, Crawford J, Narla G, Jamling TY: Sprague Dawley Rag2-Null Rats Created from Engineered Spermatogonial Stem Cells Are Immunodeficient and Permissive to Human Mol Cancer Ther.17(11): 2481-2489, 11/2018. PM30206106
  10. Belur Nagaraj A, Joseph P, Kovalenko O, Wang Q, Xu R, DiFeo A: Evaluating class III antiarrhythmic agents as novel MYC targeting drugs in ovarian cancer. Gynecol Oncol.151(3): 525-532, 12/2018. PM30301560
  11. Belur Nagaraj A, Joseph P, Ponting E, Fedorov Y, Singh S, Cole A, Lee W, Yoon E, Baccarini A, Scacheri P, Buckanovich R, Adams DJ, Drapkin R, Brown BD, DiFeo A: A miRNA-Mediated Approach to Dissect the Complexity of Tumor-Initiating Cell Function and Identify miRNA-Targeting Drugs. Stem Cell Reports.12(1): 122-134, 01/2019. PM30629937
  12. Collins G, Mesiano S, DiFeo A: Effects of Metformin on Cellular Proliferation and Steroid Hormone Receptors in Patient-Derived, Low-Grade Endometrial Cancer Cell Lines. Reprod Sci.26(5): 609-618, 05/2019. PM29848180
  13. Taylor SE, O'Connor CM, Wang Z, Shen G, Song H, Leonard D, Sangodkar J, LaVasseur C, Avril S, Waggoner S, Zanotti K, Armstrong AJ, Nagel C, Resnick K, Singh S, Jackson MW, Xu W, Haider S, DiFeo A, Narla G: The Highly Recurrent PP2A Aα-Subunit Mutation P179R Alters Protein Structure and Impairs PP2A Enzyme Function to Promote Endometrial Cancer Res.79(16): 4242-4257, 08/2019. PM31142515
  14. Shang S, Yang J, Jazaeri AA, Duval AJ, Tufan T, Lopes Fischer N, Benamar M, Guessous F, Lee I, Campbell RM, Ebert PJ, Abbas T, Landen CN, Difeo A, Scacheri PC, Adli M: Chemotherapy-Induced Distal Enhancers Drive Transcriptional Programs to Maintain the Chemoresistant State in Ovarian Cancer Res.79(18): 4599-4611, 09/2019. PM31358529
  15. Noto FK, Sangodkar J, Adedeji BT, Moody S, McClain CB, Tong M, Ostertag E, Crawford J, Gao X, Hurst L, O'Connor CM, Hanson EN, Izadmehr S, Tohmé R, Narla J, LeSueur K, Bhattacharya K, Rupani A, Tayeh MK, Innis JW, Galsky MD, Evers BM, DiFeo A, Narla G, Jamling TY: The SRG rat, a Sprague- Dawley Rag2/Il2rg double-knockout validated for human tumor oncology studies. PLoS One.15(10): e0240169, 01/2020. PM33027304
  16. Vincent J, Craig SE L, Johansen ML, Narla J, Avril S, DiFeo A, Brady-Kalnay SM: Detection of Tumor-Specific PTPmu in Gynecological Cancer and Patient Derived Diagnostics (Basel).11 (2)01/2021. PM33513911
  17. Belur Nagaraj A, Knarr M, Sekhar S, Connor RS, Joseph P, Kovalenko O, Fleming A, Surti A, Nurmemmedov E, Beltrame L, Marchini S, Kahn M, DiFeo A: The miR-181a-SFRP4 Axis Regulates Wnt Activation to Drive Stemness and Platinum Resistance in Ovarian Cancer Res.81(8): 2044-2055, 04/2021. PM33574092
  18. O'Connor CM, Taylor SE, Miller KM, Hurst L, Haanen TJ, Suhan TK, Zawacki KP, Noto FK, Trako J, Mohan A, Sangodkar J, Zamarin D, DiFeo A, Narla G: Targeting Ribonucleotide Reductase Induces Synthetic Lethality in PP2A-Deficient Uterine Serous Carcinoma. Cancer Res.82(4): 721-733, 02/2022. PM34921012
  19. Achreja A, Yu T, Mittal A, Choppara S, Animasahun O, Nenwani M, Wuchu F, Meurs N, Mohan A, Jeon JH, Sarangi I, Jayaraman A, Owen S, Kulkarni R, Cusato M, Weinberg F, Kweon HK, Subramanian C, Wicha MS, Merajver SD, Nagrath S, Cho KR, DiFeo A, Lu X, Nagrath D: Metabolic collateral lethal target identification reveals MTHFD2 paralogue dependency in ovarian cancer. Nat Metab.4(9): 1119-1137, 09/2022. PM36131208
  20. Avelar R, Armstrong A, Carvette G, Gupta R, Puleo N, Colina J, Joseph P, Sobeck A, O'Connor C, Raines B, Gandhi A, Dziubinski M, Ma D, Resnick K, Singh S, Zanotti K, Nagel C, Waggoner S, Thomas D, Skala S, Zhang J, Narla G, DiFeo A: Small molecule mediated stabilization of PP2A modulates the Homologous Recombination pathway and potentiates DNA damage-induced cell death. Mol Cancer Ther. 2023: MCT-21-0880. doi: 10.1158/1535-7163.MCT-21-0880. PMID: 36788429
  21. Kremer-Tal S, Reeves HL, Narla G, Thung SN, Schwartz M, Difeo A, Katz A, Bruix J, Bioulac-Sage P, Martignetti JA, Friedman SL: Frequent inactivation of the tumor suppressor Kruppel-like factor 6 (KLF6) in hepatocellular carcinoma. Hepatology.40(5): 1047-1052, 11/2004. PM15486921
  22. Narla G, Difeo A, Reeves HL, Schaid DJ, Hirshfeld J, Hod E, Katz A, Isaacs WB, Hebbring S, Komiya A, McDonnell SK, Wiley KE, Jacobsen SJ, Isaacs SD, Walsh PC, Zheng SL, Chang B-L, Friedrichsen DM, Stanford JL, Ostrander EA, Chinnaiyan AM, Rubin MA, Xu J, Thibodeau SN, Friedman SL, Martignetti JA: A germline DNA polymorphism enhances alternative splicing of the KLF6 tumor suppressor gene and is associated with increased prostate cancer risk. Cancer Res.65(4): 1213-1222, 02/2005. PM15735005
  23. Narla G, DiFeo A, Yao S, Banno A, Hod E, Reeves HL, Qiao RF, Camacho-Vanegas O, Levine A, Kirschenbaum A, Chan AM, Friedman SL, Martignetti JA: Targeted inhibition of the KLF6 splice variant, KLF6 SV1, suppresses prostate cancer cell growth and Cancer Res.65(13): 5761-5768, 07/2005. PM15994951
  24. DiFeo A, Narla G, Hirshfeld J, Camacho-Vanegas O, Narla J, Rose SL, Kalir T, Yao S, Levine A, Birrer MJ, Bonome T, Friedman SL, Buller RE, Martignetti JA: Roles of KLF6 and KLF6-SV1 in ovarian cancer progression and intraperitoneal dissemination. Clin Cancer Res.12(12): 3730-3739, 06/2006. PM16778100
  25. DiFeo A, Narla G, Camacho-Vanegas O, Nishio H, Rose SL, Buller RE, Friedman SL, Walsh MJ, Martignetti JA: E-cadherin is a novel transcriptional target of the KLF6 tumor Oncogene.25(44): 6026-6031, 09/2006. PM16702959
  26. Camacho-Vanegas O, Narla G, Teixeira MS, DiFeo A, Misra A, Singh G, Chan AM, Friedman SL, Feuerstein BG, Martignetti JA: Functional inactivation of the KLF6 tumor suppressor gene by loss of heterozygosity and increased alternative splicing in Int J Cancer.121(6): 1390-1395, 09/2007. PM17514651
  27. Teixeira MS, Camacho-Vanegas O, Fernandez Y, Narla G, DiFeo A, Lee B, Kalir T, Friedman SL, Schlecht NF, Genden EM, Urken M, Brandwein-Gensler M, Martignetti JA: KLF6 allelic loss is associated with tumor recurrence and markedly decreased survival in head and neck squamous cell Int J Cancer.121(9): 1976-1983, 11/2007. PM17621627
  28. DiFeo A, Feld L, Rodriguez E, Wang C, Beer DG, Martignetti JA, Narla G: A functional role for KLF6- SV1 in lung adenocarcinoma prognosis and chemotherapy Cancer Res.68(4): 965-970, 02/2008. PM18250346
  29. Narla G, DiFeo A, Fernandez Y, Dhanasekaran S, Huang F, Sangodkar J, Hod E, Leake D, Friedman SL, Hall SJ, Chinnaiyan AM, Gerald WL, Rubin MA, Martignetti JA: KLF6-SV1 overexpression accelerates human and mouse prostate cancer progression and metastasis. J Clin Invest.118(8): 2711-2721, 08/2008. PM18596922
  30. Sangodkar J, Shi J, DiFeo A, Schwartz R, Bromberg R, Choudhri A, McClinch K, Hatami R, Scheer E, Kremer-Tal S, Martignetti JA, Hui A, Leung WK, Friedman SL, Narla G: Functional role of the KLF6 tumour suppressor gene in gastric cancer. Eur J Cancer.45(4): 666-676, 03/2009. PM19101139
  31. Difeo A, Huang F, Sangodkar J, Terzo EA, Leake D, Narla G, Martignetti JA: KLF6-SV1 is a novel antiapoptotic protein that targets the BH3-only protein NOXA for degradation and whose inhibition extends survival in an ovarian cancer model. Cancer Res.69(11): 4733-4741, 06/2009. PM19435908
  32. Sangodkar J, DiFeo A, Feld L, Bromberg R, Schwartz R, Huang F, Terzo EA, Choudhri A, Narla G: Targeted reduction of KLF6-SV1 restores chemotherapy sensitivity in resistant lung Lung Cancer.66(3): 292-297, 12/2009. PM19328586
  33. Sangodkar J, Dhawan NS, Melville H, Singh VJ, Yuan E, Rana H, Izadmehr S, Farrington C, Mazhar S, Katz S, Albano T, Arnovitz P, Okrent R, Ohlmeyer M, Galsky M, Burstein D, Zhang D, Politi K, Difeo A, Narla G: Targeting the FOXO1/KLF6 axis regulates EGFR signaling and treatment response. J Clin Invest.122(7): 2637-2651, 07/2012. PM22653055
  34. Hatami R, Sieuwerts AM, Izadmehr S, Yao Z, Qiao RF, Papa L, Look MP, Smid M, Ohlssen J, Levine AC, Germain D, Burstein D, Kirschenbaum A, DiFeo A, Foekens JA, Narla G: KLF6-SV1 drives breast cancer metastasis and is associated with poor survival. Sci Transl Med.5(169): 169ra12, 01/2013. PM23345610
  35. Parikh A, Lee C, Joseph P, Marchini S, Baccarini A, Kolev V, Romualdi C, Fruscio R, Shah H, Wang F, Mullokandov G, Fishman D, D'Incalci M, Rahaman J, Kalir T, Redline RW, Brown BD, Narla G, DiFeo A: microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial- mesenchymal transition. Nat Commun.5: 2977, 01/2014. PM24394555
  36. Nagaraj AB, Joseph P, DiFeo A: miRNAs as prognostic and therapeutic tools in epithelial ovarian Biomark Med.9(3): 241-257, 01/2015. PM25731210
  37. Sangodkar J, Perl A, Tohme R, Kiselar J, Kastrinsky DB, Zaware N, Izadmehr S, Mazhar S, Wiredja DD, O'Connor CM, Hoon D, Dhawan NS, Schlatzer D, Yao S, Leonard D, Borczuk AC, Gokulrangan G, Wang L, Svenson E, Farrington CC, Yuan E, Avelar RA, Stachnik A, Smith B, Gidwani V, Giannini HM, McQuaid D, McClinch K, Wang Z, Levine AC, Sears RC, Chen EY, Duan Q, Datt M, Haider S, Ma'ayan A, DiFeo A, Sharma N, Galsky MD, Brautigan DL, Ioannou YA, Xu W, Chance MR, Ohlmeyer M, Narla G: Activation of tumor suppressor protein PP2A inhibits KRAS-driven tumor J Clin Invest.127(6): 2081- 2090, 06/2017. PM28504649
  38. McClinch K, Avelar RA, Callejas D, Izadmehr S, Wiredja D, Perl A, Sangodkar J, Kastrinsky DB, Schlatzer D, Cooper M, Kiselar J, Stachnik A, Yao S, Hoon D, McQuaid D, Zaware N, Gong Y, Brautigan DL, Plymate SR, Sprenger CC T, Oh WK, Levine AC, Kirschenbaum A, Sfakianos JP, Sears R, DiFeo A, Ioannou Y, Ohlmeyer M, Narla G, Galsky MD: Small-Molecule Activators of Protein Phosphatase 2A for the Treatment of Castration-Resistant Prostate Cancer. Cancer Res.78(8): 2065-2080, 04/2018. PM29358171
  39. Parasido E, Avetian GS, Naeem A, Graham G, Pishvaian M, Glasgow E, Mudambi S, Lee Y, Ihemelandu C, Choudhry M, Peran I, Banerjee PP, Avantaggiati ML, Bryant K, Baldelli E, Pierobon M, Liotta L, Petricoin E, Fricke ST, Sebastian A, Cozzitorto J, Loots GG, Kumar D, Byers S, Londin E, DiFeo A, Narla G, Winter J, Brody JR, Rodriguez O, Albanese C: The Sustained Induction of c-MYC Drives Nab-Paclitaxel Resistance in Primary Pancreatic Ductal Carcinoma Cells. Mol Cancer Res.17(9): 1815-1827, 09/2019. PM31164413