Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Filemon Dela Cruz is active.

Publication


Featured researches published by Filemon Dela Cruz.


Oncogene | 2013

Terminal differentiation and loss of tumorigenicity of human cancers via pluripotency based reprogramming

Xi Zhang; Filemon Dela Cruz; Melissa Terry; Fabrizio Remotti; Igor Matushansky

Pluripotent cells can be derived from various types of somatic cells by nuclear reprogramming using defined transcription factors. It is, however, unclear whether human cancer cells can be similarly reprogrammed and subsequently terminally differentiated with abrogation of tumorigenicity. Here, using sarcomas we show that human-derived complex karyotype solid tumors: (1) can be reprogrammed into a pluripotent-like state as defined by all in vitro criteria used to define pluripotent stem cells generated from somatic cells; (2) can be terminally differentiated into mature connective tissue and red blood cells; and (3) terminal differentiation is accompanied with loss of both proliferation and tumorigenicity. We go on to perform the first global DNA promoter methylation and gene expression analyses comparing human cancers to their reprogrammed counterparts and report that reprogramming/differentiation results in significant epigenetic remodeling of oncogenes and tumor suppressors, while not significantly altering the differentiation status of the reprogrammed cancer cells, in essence dedifferentiating them to a state slightly before the mesenchymal stem cell differentiation stage. Our data demonstrate that direct nuclear reprogramming can restore terminal differentiation potential to human-derived cancer cells, with simultaneous loss of tumorigenicity, without the need to revert to an embryonic state. We anticipate that our models would serve as a starting point to more fully assess how nuclear reprogramming overcomes the multitude of genetic and epigenetic aberrancies inherent in human cancers to restore normal terminal differentiation pathways. Finally, these findings suggest that nuclear reprogramming may be a broadly applicable therapeutic strategy for the treatment of cancer.


Genome Medicine | 2016

Implementation of next generation sequencing into pediatric hematology-oncology practice: moving beyond actionable alterations

Jennifer A. Oberg; Julia L. Glade Bender; Maria Luisa Sulis; Danielle Pendrick; Anthony N. Sireci; Susan J. Hsiao; Andrew T. Turk; Filemon Dela Cruz; Hanina Hibshoosh; Helen Remotti; Rebecca J. Zylber; Jiuhong Pang; Daniel Diolaiti; Stuart J. Andrews; James Garvin; Darrell J. Yamashiro; Wendy K. Chung; Stephen G. Emerson; Peter L. Nagy; Mahesh Mansukhani; Andrew L. Kung

BackgroundMolecular characterization has the potential to advance the management of pediatric cancer and high-risk hematologic disease. The clinical integration of genome sequencing into standard clinical practice has been limited and the potential utility of genome sequencing to identify clinically impactful information beyond targetable alterations has been underestimated.MethodsThe Precision in Pediatric Sequencing (PIPseq) Program at Columbia University Medical Center instituted prospective clinical next generation sequencing (NGS) for pediatric cancer and hematologic disorders at risk for treatment failure. We performed cancer whole exome sequencing (WES) of patient-matched tumor-normal samples and RNA sequencing (RNA-seq) of tumor to identify sequence variants, fusion transcripts, relative gene expression, and copy number variation (CNV). A directed cancer gene panel assay was used when sample adequacy was a concern. Constitutional WES of patients and parents was performed when a constitutionally encoded disease was suspected. Results were initially reviewed by a molecular pathologist and subsequently by a multi-disciplinary molecular tumor board. Clinical reports were issued to the ordering physician and posted to the patient’s electronic medical record.ResultsNGS was performed on tumor and/or normal tissue from 101 high-risk pediatric patients. Potentially actionable alterations were identified in 38% of patients, of which only 16% subsequently received matched therapy. In an additional 38% of patients, the genomic data provided clinically relevant information of diagnostic, prognostic, or pharmacogenomic significance. RNA-seq was clinically impactful in 37/65 patients (57%) providing diagnostic and/or prognostic information for 17 patients (26%) and identified therapeutic targets in 15 patients (23%). Known or likely pathogenic germline alterations were discovered in 18/90 patients (20%) with 14% having germline alternations in cancer predisposition genes. American College of Medical Genetics (ACMG) secondary findings were identified in six patients.ConclusionsOur results demonstrate the feasibility of incorporating clinical NGS into pediatric hematology-oncology practice. Beyond the identification of actionable alterations, the ability to avoid ineffective/inappropriate therapies, make a definitive diagnosis, and identify pharmacogenomic modifiers is clinically impactful. Taking a more inclusive view of potential clinical utility, 66% of cases tested through our program had clinically impactful findings and samples interrogated with both WES and RNA-seq resulted in data that impacted clinical decisions in 75% of cases.


Cold Spring Harb Mol Case Stud | 2015

Characterization of a novel fusion gene EML4-NTRK3 in a case of recurrent congenital fibrosarcoma

Sarah Tannenbaum-Dvir; Julia L. Glade Bender; Alanna Church; Katherine A. Janeway; Marian H. Harris; Mahesh Mansukhani; Peter L. Nagy; Stuart J. Andrews; Vundavalli V. Murty; Angela Kadenhe-Chiweshe; E.P. Connolly; Andrew L. Kung; Filemon Dela Cruz

Abstract We describe the clinical course of a recurrent case of congenital fibrosarcoma diagnosed in a 9-mo-old boy with a history of hemimelia. Following complete surgical resection of the primary tumor, the patient subsequently presented with bulky bilateral pulmonary metastases 6 mo following surgery. Molecular characterization of the tumor revealed the absence of the prototypical ETV6-NTRK3 translocation. However, tumor characterization incorporating cytogenetic, array comparative genomic hybridization, and RNA sequencing analyses, revealed a somatic t(2;15)(2p21;15q25) translocation resulting in the novel fusion of EML4 with NTRK3. Cloning and expression of EML4-NTRK3 in murine fibroblast NIH 3T3 cells revealed a potent tumorigenic phenotype as assessed in vitro and in vivo. These results demonstrate that multiple fusion partners targeting NTRK3 can contribute to the development of congenital fibrosarcoma.


Modern Pathology | 2018

Recurrent EML4–NTRK3 fusions in infantile fibrosarcoma and congenital mesoblastic nephroma suggest a revised testing strategy

Alanna Church; Monica L. Calicchio; Valentina Nardi; Alena Skálová; Andre Pinto; Deborah A. Dillon; Carmen R Gomez-Fernandez; Namitha Manoj; Josh D Haimes; Joshua A Stahl; Filemon Dela Cruz; Sarah Tannenbaum-Dvir; Julia Glade-Bender; Andrew L. Kung; Steven G. DuBois; Harry P. Kozakewich; Katherine A. Janeway; Antonio R. Perez-Atayde; Marian H. Harris

Infantile fibrosarcoma and congenital mesoblastic nephroma are tumors of infancy traditionally associated with the ETV6–NTRK3 gene fusion. However, a number of case reports have identified variant fusions in these tumors. In order to assess the frequency of variant NTRK3 fusions, and in particular whether the recently identified EML4–NTRK3 fusion is recurrent, 63 archival cases of infantile fibrosarcoma, congenital mesoblastic nephroma, mammary analog secretory carcinoma and secretory breast carcinoma (tumor types that are known to carry recurrent ETV6–NTRK3 fusions) were tested with NTRK3 break-apart FISH, EML4–NTRK3 dual fusion FISH, and targeted RNA sequencing. The EML4–NTRK3 fusion was identified in two cases of infantile fibrosarcoma (one of which was previously described), and in one case of congenital mesoblastic nephroma, demonstrating that the EML4–NTRK3 fusion is a recurrent genetic event in these related tumors. The growing spectrum of gene fusions associated with infantile fibrosarcoma and congenital mesoblastic nephroma along with the recent availability of targeted therapies directed toward inhibition of NTRK signaling argue for alternate testing strategies beyond ETV6 break-apart FISH. The use of either NTRK3 FISH or next-generation sequencing will expand the number of cases in which an oncogenic fusion is identified and facilitate optimal diagnosis and treatment for patients.


Molecular Cancer Therapeutics | 2017

Inhibition of Hsp90 Suppresses PI3K/AKT/mTOR Signaling and Has Antitumor Activity in Burkitt Lymphoma

Lisa Giulino-Roth; Herman van Besien; Tanner Dalton; Jennifer Totonchy; Anna Rodina; Tony Taldone; Alexander Bolaender; Hediye Erdjument-Bromage; Jouliana Sadek; Amy Chadburn; Matthew J. Barth; Filemon Dela Cruz; Allison R. Rainey; Andrew L. Kung; Gabriela Chiosis; Ethel Cesarman

Hsp90 is a molecular chaperone that protects proteins, including oncogenic signaling complexes, from proteolytic degradation. PU-H71 is a next-generation Hsp90 inhibitor that preferentially targets the functionally distinct pool of Hsp90 present in tumor cells. Tumors that are driven by the MYC oncoprotein may be particularly sensitive to PU-H71 due to the essential role of Hsp90 in the epichaperome, which maintains the malignant phenotype in the setting of MYC. Burkitt lymphoma (BL) is an aggressive B-cell lymphoma characterized by MYC dysregulation. In this study, we evaluated Hsp90 as a potential therapeutic target in BL. We found that primary BL tumors overexpress Hsp90 and that Hsp90 inhibition has antitumor activity in vitro and in vivo, including potent activity in a patient-derived xenograft model of BL. To evaluate the targets of PU-H71 in BL, we performed high-affinity capture followed by proteomic analysis using mass spectrometry. We found that Hsp90 inhibition targets multiple components of PI3K/AKT/mTOR signaling, highlighting the importance of this pathway in BL. Finally, we found that the anti-lymphoma activity of PU-H71 is synergistic with dual PI3K/mTOR inhibition in vitro and in vivo. Overall, this work provides support for Hsp90 as a therapeutic target in BL and suggests the potential for combination therapy with PU-H71 and inhibitors of PI3K/mTOR. Mol Cancer Ther; 16(9); 1779–90. ©2017 AACR.


Science Translational Medicine | 2017

Therapeutic targeting of PGBD5-induced DNA repair dependency in pediatric solid tumors

Anton Henssen; Casie Reed; Eileen Jiang; Heathcliff Dorado García; Jennifer von Stebut; Ian Macarthur; Patrick Hundsdoerfer; Jun Hyun Kim; Elisa de Stanchina; Yasumichi Kuwahara; Hajime Hosoi; Neil J. Ganem; Filemon Dela Cruz; Andrew L. Kung; Johannes H. Schulte; John H.J. Petrini; Alex Kentsis

PGBD5 DNA transposase confers therapeutically actionable dependency in solid tumors. Synthetic lethality, pediatric edition Although a variety of therapeutic regimens are available for pediatric solid tumors, they are often ineffective and typically nonspecific. Henssen et al. determined that expression of a DNA transposase called PGBD5 is common in these tumors and presents a therapeutic vulnerability. The authors demonstrated that cells expressing PGBD5 are dependent on DNA repair through nonhomologous end joining, then identified a drug that inhibits this DNA repair pathway and is therefore active against many pediatric tumor types, particularly when combined with chemotherapy, while sparing surrounding nontumor tissues. Despite intense efforts, the cure rates of childhood and adult solid tumors are not satisfactory. Resistance to intensive chemotherapy is common, and targets for molecular therapies are largely undefined. We have found that the majority of childhood solid tumors, including rhabdoid tumors, neuroblastoma, medulloblastoma, and Ewing sarcoma, express an active DNA transposase, PGBD5, that can promote site-specific genomic rearrangements in human cells. Using functional genetic approaches, we discovered that mouse and human cells deficient in nonhomologous end joining (NHEJ) DNA repair cannot tolerate the expression of PGBD5. In a chemical screen of DNA damage signaling inhibitors, we identified AZD6738 as a specific sensitizer of PGBD5-dependent DNA damage and apoptosis. We found that expression of PGBD5, but not its nuclease activity–deficient mutant, was sufficient to induce sensitivity to AZD6738. Depletion of endogenous PGBD5 conferred resistance to AZD6738 in human tumor cells. PGBD5-expressing tumor cells accumulated unrepaired DNA damage in response to AZD6738 treatment and underwent apoptosis in both dividing and G1-phase cells in the absence of immediate DNA replication stress. Accordingly, AZD6738 exhibited nanomolar potency against most neuroblastoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumor cells tested while sparing nontransformed human and mouse embryonic fibroblasts in vitro. Finally, treatment with AZD6738 induced apoptosis and regression of human neuroblastoma and medulloblastoma tumors engrafted in immunodeficient mice in vivo. This effect was potentiated by combined treatment with cisplatin, including substantial antitumor activity against patient-derived primary neuroblastoma xenografts. These findings delineate a therapeutically actionable synthetic dependency induced in PGBD5-expressing solid tumors.


Genome Medicine | 2016

A case study of an integrative genomic and experimental therapeutic approach for rare tumors: identification of vulnerabilities in a pediatric poorly differentiated carcinoma.

Filemon Dela Cruz; Daniel Diolaiti; Andrew T. Turk; Allison R. Rainey; Alberto Ambesi-Impiombato; Stuart J. Andrews; Mahesh Mansukhani; Peter L. Nagy; Mariano J. Alvarez; Farhad Forouhar; Beata Modzelewski; Chelsey M. Mitchell; Darrell J. Yamashiro; Lianna Marks; Julia L. Glade Bender; Andrew L. Kung

BackgroundPrecision medicine approaches are ideally suited for rare tumors where comprehensive characterization may have diagnostic, prognostic, and therapeutic value. We describe the clinical case and molecular characterization of an adolescent with metastatic poorly differentiated carcinoma (PDC). Given the rarity and poor prognosis associated with PDC in children, we utilized genomic analysis and preclinical models to validate oncogenic drivers and identify molecular vulnerabilities.MethodsWe utilized whole exome sequencing (WES) and transcriptome analysis to identify germline and somatic alterations in the patient’s tumor. In silico and in vitro studies were used to determine the functional consequences of genomic alterations. Primary tumor was used to generate a patient-derived xenograft (PDX) model, which was used for in vivo assessment of predicted therapeutic options.ResultsWES revealed a novel germline frameshift variant (p.E1554fs) in APC, establishing a diagnosis of Gardner syndrome, along with a somatic nonsense (p.R790*) APC mutation in the tumor. Somatic mutations in TP53, MAX, BRAF, ROS1, and RPTOR were also identified and transcriptome and immunohistochemical analyses suggested hyperactivation of the Wnt/ß-catenin and AKT/mTOR pathways. In silico and biochemical assays demonstrated that the MAX p.R60Q and BRAF p.K483E mutations were activating mutations, whereas the ROS1 and RPTOR mutations were of lower utility for therapeutic targeting. Utilizing a patient-specific PDX model, we demonstrated in vivo activity of mTOR inhibition with temsirolimus and partial response to inhibition of MEK.ConclusionsThis clinical case illustrates the depth of investigation necessary to fully characterize the functional significance of the breadth of alterations identified through genomic analysis.


Cancer Investigation | 2013

Chemotherapy Use in Elderly Patients with Soft Tissue Sarcoma: A Population-based Study

Igor Matushansky; Filemon Dela Cruz; Beverly J. Insel; Dawn L. Hershman; Alfred I. Neugut

Adjuvant chemotherapy for soft tissue sarcoma (STS) remains controversial while improvement in survival has never been conclusively demonstrated for metastatic STS. We identified individuals in SEER–Medicare with resected or metastatic STS, 1991–2007. Of 2,382 patients with resected STS, 106 (4.5%) received chemotherapy. High tumor grade, larger tumor size, and malignant fibrous histiocytoma subtype were associated with chemotherapy receipt. Of 365 patients with metastatic STS, 118 (32.4%) received chemotherapy. Younger age, fewer comorbidities, and being married were associated with chemotherapy receipt. Consistent with clinical trials, we found minimal use of chemotherapy. Clinical factors were associated with chemotherapy receipt in nonmetastatic STS.


Differentiation | 2012

A transgenic, mesodermal specific, Dkk1 mouse model recapitulates a spectrum of human congenital limb reduction defects.

Filemon Dela Cruz; Melissa Terry; Igor Matushansky

Congenital limb reduction defects occurring in isolation of other developmental abnormalities continue to be an important medical problem in which little progress has been made. Herein we generated transgenic mice expressing Dkk1 in an appendicular mesodermal pattern. Prx1-Dkk1 mice recapitulate a full spectrum of human congenital limb reduction defects, without other developmental issues, and have normal life-spans. Importantly, a close examination of the inheritance pattern suggests that there is a significant degree of incomplete penetrance as progeny of phenotypically positive or phenotypically negative, but genotypically positive Prx1-Dkk1 mice, consistently give rise to both phenotypically positive mice and phenotypically normal-appearing mice. Thus, this heterogeneous phenotype is reproducible with each generation regardless of the phenotype of the parents. We further go on to identify that mesenchymal stem cells from Prx1-Dkk1 mice have limited proliferative ability, but normal differentiation potential, which may explain the mechanism for the limb reduction defects observed. We believe Prx1-Dkk1 mice may prove useful in the future to study the mechanisms underlying the development of congenital limb reduction defects.


Nature Genetics | 2018

A precision oncology approach to the pharmacological targeting of mechanistic dependencies in neuroendocrine tumors

Mariano J. Alvarez; Prem S. Subramaniam; Laura H. Tang; Adina Grunn; Mahalaxmi Aburi; Gabrielle E. Rieckhof; Elena V. Komissarova; Elizabeth A. Hagan; Lisa Bodei; Paul A. Clemons; Filemon Dela Cruz; Deepti Dhall; Daniel Diolaiti; Douglas A. Fraker; Afshin Ghavami; Daniel Kaemmerer; Charles Karan; Mark Kidd; Kyoung Mee Kim; Hee C. Kim; Lakshmi P. Kunju; Ülo Langel; Zhong Li; Jeeyun Lee; Hai Li; Virginia A. LiVolsi; Roswitha Pfragner; Allison R. Rainey; Ronald Realubit; Helen Remotti

We introduce and validate a new precision oncology framework for the systematic prioritization of drugs targeting mechanistic tumor dependencies in individual patients. Compounds are prioritized on the basis of their ability to invert the concerted activity of master regulator proteins that mechanistically regulate tumor cell state, as assessed from systematic drug perturbation assays. We validated the approach on a cohort of 212 gastroenteropancreatic neuroendocrine tumors (GEP-NETs), a rare malignancy originating in the pancreas and gastrointestinal tract. The analysis identified several master regulator proteins, including key regulators of neuroendocrine lineage progenitor state and immunoevasion, whose role as critical tumor dependencies was experimentally confirmed. Transcriptome analysis of GEP-NET-derived cells, perturbed with a library of 107 compounds, identified the HDAC class I inhibitor entinostat as a potent inhibitor of master regulator activity for 42% of metastatic GEP-NET patients, abrogating tumor growth in vivo. This approach may thus complement current efforts in precision oncology.This study presents OncoTreat, a framework for the prioritization of compounds targeting mechanistic tumor dependencies in individual patients.

Collaboration


Dive into the Filemon Dela Cruz's collaboration.

Top Co-Authors

Avatar

Andrew L. Kung

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Diolaiti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Igor Matushansky

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Allison R. Rainey

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ervin Gaviria

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia L. Glade Bender

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mahesh Mansukhani

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge