Joseph R. Testa, PhD, FACMG

Joseph Testa, PhD
​​

This Fox Chase professor participates in the Undergraduate Summer Research Fellowship
Learn more about Research Volunteering.

Professor/Senior Member

Carol & Kenneth E. Weg Chair in Human Genetics

Chief, Genomic Medicine

Director, Clinical Cytogenomics Laboratory

  • Figure 1. Incidence and latency of pleural malignant mesothelioma (MM) in mouse cohorts with different genotypes following intrathoracic (IT) injections with Adeno-Cre virus. A, Incidence of MM in the various conditional knockout mouse (CKO) mouse groups. B, Tumor latency curves for MMs arising after Adeno-Cre injections in mice with CKO of different combinations of Bap1, Nf2 and/or Cdkn2a tumor suppressor genes. Kaplan-Meier curves depict survival of mice succumbing to MM arising after IT injections with Adeno-Cre. Note rapid development of MM in triple-CKO mice, with the median survival being only 12 weeks, which was significantly different (P < 0.0001) from that of any of the other compound CKO mice.

  • Figure 2. Treatment of human malignant mesothelioma (MM) cells with DNA methylation and/or histone deacetylase inhibitors results in restoration of RIPK3 expression. A-C, The expression of RIPK3 was also determined after treatment of MM cell lines with 2.5, 10 or 30 µM 5-aza-2’-deoxycyditine (5-Aza) alone for 7 days or with 2.5, 10 or 30 µM 5-Aza for 6 days with TSA for the final 24 h (7 days total), followed by semi-quantitative RT-PCR (A), Real-Time PCR (B) and immunoblot analysis (C). D, RIPK3-positive cell lines (M29 and M34) and RIPK3-negative cell lines (M12 and M20) were used to study DNA methylation. Genomic DNA fragments with methylated-CpG were enriched with MBD2b/MBD3L1 and subjected to NGS and analyzed using MACS2 peak calling pipeline. Both RIPK3-negative cell lines show strong methylated-CpG peaks in RIPK3 intron1 and exon2, whereas both RIPK3-positive cell lines do not. The pre-captured DNAs serve as negative controls (Input). Histograph depicts fold changes of the methylated-CpG peaks among the cell lines studied.

  • Figure 3. RIPK3 suppresses clonogenic growth of MM cells. A, RIPK3-negative cells were nucleofected with pEGFP, pEGFP-RIPK3-WT or pEGFP-RIPK3-KD, and clonogenic assays were then performed. B, Quantification of colonies was performed in triplicate with error bars indicated. C, Immunoblot analysis of RIPK3, p-MLKL, MLKL and GAPDH in nucleofected MM cells. The cells used for immunoblotting in panel C were harvested 48 h after nucleofection; and the remaining cells were seeded on dishes and selected in medium containing G418 and allowed to form the colonies shown in panel B.

  • Figure 4. Myristylated Akt2 (MyrAkt2) cooperates with the homeobox protein Dlx5 to accelerate T-cell lymphomagenesis. (A) Survival curves of Lck-MyrAkt2, Lck-Dlx5, and Lck-Dlx5;Lck-MyrAkt2 transgenic mice dying due to T-ALL. The number of animals for each genotype was as follows: Lck-MyrAkt2: 20 mice, Lck-Dlx5: 40 mice, and Lck-Dlx5;Lck-MyrAkt2: 53 mice. (B) H&E staining depicting T-ALL infiltration and dissemination in an Lck-Dlx5;Lck-MyrAkt2 mouse. (C) Immunoblot demonstrating expression of Notch1, Notch3, Myc-tagged Dlx5, Lef1, β-catenin, and c-Myc in lymphomas from Lck-MyrAkt2, Lck-Dlx5, and Lck-Dlx5;Lck-MyrAkt2 mice compared to that of normal thymic T cells from wild-type (WT) mice.

  • Chromosomal rearrangements in mesothelioma cells from a p19Arf-deficient mouse

  • Pedigrees of two U.S. families with a high incidence of mesothelioma and other cancers.

  • Homozygous losses of the CDKN2A/ARF locus in human mesothelioma

    Homozygous losses of the CDKN2A/ARF locus in human mesothelioma

  • Effect of mTOR inhibition in a mouse model of ovarian cancer

  • Genomic copy number analysis profile

  • Immunohistochemical staining for phosphorylated PAK

    Immunohistochemical staining for phosphorylated PAK (p21-acivated kinase)

  • Analysis of recurrent chromosome 6 inversion.

  • Testa Research Lab. Mitchell Cheung, Ph.D. (left), Ujjawal Shrestha, M.D. (second from left), Yuwaraj Kadariya (third from left), and Eleonora Sementino , M.S. (right).

     

    Educational Background

    • Postdoctorate, University of Chicago, Chicago, IL, 1976-1980
    • PhD, Biological Sciences, Fordham University, New York, NY, 1976
    • BS, MS, Biology, Southern Connecticut State University, New Haven, CT, 1973

     

    Certifications

    • Diplomate (Clinical Cytogenetics), American Board of Medical Genetics, 1987
    • Founding Fellow PhD, American College of Medical Genetics & Genomics (FACMG), 1993

    Memberships

    • American Association for the Advancement of Science
    • American Association for Cancer Research
    • American College of Medical Genetics
    • American Society of Hematology
    • American Society of Human Genetics
    • International Mesothelioma Interest Group

    Honors & Awards

    • AACR Most-Cited Article Published in 2016 – Cancer Prevention Research, 2018
    • Pioneer Research Award, Mesothelioma Applied Research Foundation, 2013
    • Wagner Medal, International Mesothelioma Interest Group, 2012
    • Elected Fellow, American Association for the Advancement of Science, 2012
    • The Reimann Honor Award of the Fox Chase Cancer Center, 2011
    • Scientific Research Award, American Cancer Society (ACS) Southeast PA Division, 2009
    • Co-recipient, Landon Foundation-AACR Innovator Award for International Collaboration in Cancer Research (team award), 2008
    • Member, NCI Board of Scientific Counselors, Basic Sciences, 2006-2011
    • Irving Selikoff Award for Cancer Research, 1999
    • Stohlman Memorial Scholar Award, Leukemia Society, 1987
    • Leukemia Society of America Scholar Award, 1984-1990
    • Leukemia Society of America Special Fellow Award, 1982-1984

    People

    Research Facility

    Research Interests

    Molecular biology of mesothelioma; role of AKT in oncogenesis

    • The BAP1 tumor predisposition syndrome and other genetic factors that may predispose to mesothelioma
    • Molecular genetic basis of malignant mesothelioma susceptibility and progression.
    • Role of the AKT1/2 oncogenes in tumorigenesis and resistance to anti-cancer therapies.
    • Characterization of genes that interact with and/or cooperate with AKT kinase activation in tumor development.
    • Use of genetically engineered mice to assess gene–environment interactions, the role of asbestos-induced inflammation in tumor development, and as preclinical models for selective drug targeting of cellular pathways underpinning human tumorigenesis.

    Lab Overview

    The Testa lab focuses on two research areas: 1) hereditary and somatic genetics of malignant mesothelioma, a cancer of the serosal cells lining the chest and abdominal cavities, primarily caused by exposure to asbestos; and 2) the role of AKT oncogenes in tumorigenesis.  The lab integrates data from human primary tumor tissues and tumor-derived cell lines as well as tumors from genetically engineered mouse models (GEMM) to address research questions in these two subject areas.  As a result of the group’s interest in hereditary aspects of mesothelioma, the lab discovered the first germline (heritable) BAP1 mutations in two families with a high incidence of mesothelioma, and the group observed somatic alterations affecting BAP1 in familial mesotheliomas, indicating biallelic inactivation. In addition to mesothelioma, some BAP1 mutation carriers developed uveal melanoma or other cancers, consistent with the existence of a novel tumor syndrome, now known as BAP1 tumor predisposition syndrome (BAP1-TPDS).  The group is also investigating other genes that may contribute to mesothelioma susceptibility and progression.  GEMM are being used to assess gene–environment interactions in mesothelioma pathogenesis and as preclinical models for targeting cellular pathways important in this disease.  With regard to AKT, the lab continues to study the role of the AKT1 and AKT2 oncogenes in tumorigenesis and drug resistance to therapies.

    Lab Description

    The Testa lab investigates the role of hereditary and somatic mutations in malignant mesothelioma, an incurable form of cancer often caused by exposure to asbestos.  The group discovered frequent mutations of the CDKN2A locus, a region of DNA that encodes the tumor suppressors p16INK4A and p14ARF, and NF2/merlin in human mesothelioma.  In 2011, he and his collaborators also discovered germline mutations of the BAP1 tumor suppressor gene in families with a high incidence of mesothelioma, the first study demonstrating that inherited mutations can influence a person’s risk of mesothelioma.  Besides mesothelioma, some of the BAP1 mutation carriers developed ocular melanoma or other cancers, and this cancer susceptibility is now recognized as the BAP1 tumor predisposition syndrome (BAP1-TPDS).  The Testa lab has reported multiple families with the BAP1-TPDS, including some family members with two or more different types of cancer, suggesting widespread BAP1-related tumor susceptibility targeting tissues of multiple organs.  One of the families with a germline nonsense mutation in BAP1 included five relatives with peritoneal mesotheliomas as well as second or third primary cancers, including two with meningiomas. Two family members had basal cell carcinomas, and six others had melanocytic tumors, including four cutaneous melanomas, one uveal melanoma, and one benign melanocytic tumor. Given that this family resides in a subtropical area, and several members had suspected exposure to asbestos either occupationally or in the home, we hypothesized that the concurrence of a genetic predisposing factor and environmental exposure to asbestos and UV irradiation contributed to the high incidence of multiple cancers seen in this family, specifically mesothelioma and various uveal/skin tumors, respectively.

    In another, large study, the Testa lab examined the germline BAP1 mutation status of 150 mesothelioma patients with a family history of cancer, 50 asbestos-exposed control individuals with a family history of cancers other than mesothelioma, and 153 asbestos-exposed individuals without familial cancer. No BAP1 alterations were found in control cohorts, but were identified in 9 of 150 mesothelioma cases (6%) with a family history of cancer. Alterations among these cases were characterized by both missense and frameshift mutations, and enzymatic activity of BAP1 missense mutants was decreased compared with wild-type BAP1. Furthermore, BAP1 mutation carriers developed mesothelioma at an earlier age that was more often peritoneal than pleural (5 of 9) and exhibited improved long-term survival compared to mesothelioma patients without BAP1 mutations. Moreover, many tumors harboring BAP1 germline mutations were associated with the BAP1-TPDS. Collectively, these findings suggest that mesothelioma patients presenting with a family history of cancer should be considered for BAP1 genetic testing to identify those individuals who might benefit from further screening and routine monitoring for the purpose of early detection and intervention.  In a subsequent international intergroup study that included the Testa lab, 181 BAP1-TPDS families worldwide were reviewed, and the collated data confirmed the core tumor spectrum associated with the syndrome.  Median ages of onset of core tumor types were lower in null than missense variant carriers for all tumors combined, mesothelioma, cutaneous melanoma, and nonmelanoma skin cancer (each having P values of < 0.001).  In addition to BAP1, the Testa lab is also investigating other genes that may predispose to mesothelioma.

    Much of the current work in the lab is focused understanding the mechanisms involved in BAP1-related tumor susceptibility.  Using several GEMM they developed, the Testa lab provided the first in vivo evidence that heritable (germline) mutations of Bap1 predispose to the development of asbestos-induced mesothelioma. This and other Bap1-mutant mouse models are currently being used to assess susceptibility to spontaneous mesothelioma formation and  gene–environment interactions.  In one project, the Testa lab introduced various combinations of deletions of Bap1, Cdkn2a, and/or Nf2 in the pleura of conditional knockout (CKO) mice, focusing on the contribution of Bap1 loss. While homozygous CKO of Bap1, Cdkn2a, or Nf2 alone gave rise to few or no mesotheliomas, inactivation of Bap1 cooperated with loss of either Nf2 or Cdkn2a to drive development of mesothelioma in approximately 20% of double-CKO mice, and a high incidence (22/26, 85%) of mesotheliomas was observed in Bap1;Nf2;Cdkn2a (triple)-CKO mice. Malignant mesothelioma onset was rapid in triple-CKO mice, with a median survival of only 12 weeks, and the mesotheliomas from these mice were consistently high-grade and invasive. Adenoviral-Cre treatment of normal mesothelial cells from Bap1;Nf2;Cdkn2a CKO mice, but not from mice with knockout of one or any two of these genes, resulted in robust spheroid formation in vitro, suggesting that mesothelial cells from Bap1;Nf2;Cdkn2a mice have stem cell-like potential. RNA-seq analysis of mesotheliomas from triple-CKO mice revealed enrichment of genes transcriptionally regulated by the polycomb repressive complex 2 (PRC2) and others previously implicated in known Bap1-related cellular processes. These data demonstrate that somatic inactivation of Bap1, Nf2, and Cdkn2a results in rapid, aggressive mesotheliomas, and that deletion of Bap1 contributes to tumor development, in part, by loss of PRC2-mediated repression of tumorigenic target genes and by acquisition of stem cell potential, suggesting a potential avenue for therapeutic intervention.

    The group is also using novel GEMM to unravel the role of asbestos-induced inflammation in the genesis of malignant mesothelioma.  For example, the Testa lab demonstrated that inflammation-related IL1β/IL1R signaling promotes the onset of asbestos-induced malignant mesothelioma, and the team’s in vivo findings provided rationale for chemoprevention strategies targeting IL1β/IL1R signaling in high-risk, asbestos-exposed populations. Ongoing preclinical studies with mouse models are being performed to assess the efficacy of certain other anti-inflammatory drugs as chemoprevention agents.

    Mechanistic work by the Testa lab previously linked NF2 inactivation to oncogenic PAK and FAK signaling, implicating NF2 inactivation in mesothelioma cell spreading, invasiveness and proliferation, thereby establishing a framework for elucidating tumorigenic mechanisms and novel therapeutic targets in this disease.   Other in vivo studies demonstrated that genetically engineered deletions of Nf2 and Cdkn2a cooperate to drive the development of highly aggressive mesotheliomas characterized by enhanced tumor dissemination and the involvement of a cancer stem cell (CSC) population.  Mesothelioma is very difficult to treat and almost always recurs after therapy.  Based on his work linking NF2 loss to FAK activation, he partnered with investigators in the pharmaceutical industry to test a novel drug that blocks FAK activity.  Their findings suggested that FAK inhibitor treatment might be especially beneficial in patients with NF2-deficient tumors.  Moreover, the drug proved to be particularly effective at killing CSCs, which can give rise to recurrent tumors.  These preclinical studies provided the rationale for a clinical trial in mesothelioma patients using a FAK inhibitor as a single agent after first-line chemotherapy. More recently, other mechanistic studies have demonstrated that RIPK3, which encodes a receptor-interacting protein kinase, acts as a tumor suppressor in mesothelioma by triggering necroptosis, and that epigenetic silencing of RIPK3 by DNA methylation inactivates necroptosis and contributes to chemoresistance and poor survival in this incurable disease.

    Testa has had a longstanding interest in the oncogenic role of AKT, beginning with his chromosomal mapping of the AKT1 proto-oncogene in 1988.  The following year, Testa brought the AKT project to Fox Chase, when he joined the Center and jointly published a series of seminal studies with Phil Tsichlis and Alfonso Bellacosa.  In a highly cited 1991 paper in Science, the team reported that the retroviral oncogene, akt, encodes a predicted oncoprotein contained viral Gag sequences fused to a serine-threonine kinase related to protein kinase C. The oncogenic potential of v-akt arises from the creation of a myristylation site at the amino terminus and consequent constitutive kinase activity, whereas the cellular homolog c-akt requires mitogenic stimulation for its activation.  The Testa lab cloned and characterized the related AKT2 gene and provided the first evidence for recurrent alterations of the AKT pathway in human cancers.  In recent years, the lab has characterized how other genes, such as the proto-oncogene Myc or the homeobox gene Dlx5 cooperate with Akt2 to promote T-cell lymphomagenesis.  Recently, the Testa lab generated compound Dlx5;Akt2 transgenic mice, and the team discovered that activated Akt2 synergized with over-expressed Dlx5 to greatly accelerate and enhance the dissemination of T-lymphomagenesis. RNA-seq analysis performed on lymphomas from these mice revealed upregulation of genes involved in the Wnt and cholesterol biosynthesis pathways. Combined RNA-seq and ChIP-seq analysis of lymphomas from Dlx5;Akt2 mice demonstrated that β-catenin directly regulates genes involved in sterol regulatory element binding transcription factor 2 (Srebf2)-cholesterol synthesis. These lymphoma cells had high Lef1 levels and were highly sensitive to β-catenin and Srebf2-cholesterol synthesis inhibitors. Similarly, human T-cell leukemia/lymphoma (T-ALL) cell lines with activated NOTCH and AKT and elevated LEF1 levels were sensitive to inhibition of β-catenin and cholesterol pathways. Furthermore, LEF1 expression positively correlated with expression of genes involved in the cholesterol synthesis pathway in primary human T-ALL specimens. Together, these data suggest that targeting β-catenin and/or cholesterol biosynthesis, together with AKT, could have therapeutic efficacy in a subset of T-ALL patients.

    Misc

    Extramural Affiliations

    University of Pennsylvania

    Selected Publications

    Krais J., Glass D., Chudoba I., Wang Y., Feng W., Simpson D., Patel P., Liu Z., Neumann-Domer R., Betsch R., Bernhardy A., Bradbury A., Conger J., Yueh W.-T., Nacson J., Pomerantz R., Gupta G., Testa J.R., Johnson N. Genetic separation of Brca1 functions reveal pre-mitotic RPA as a driver of Polq addiction. Nat Commun. (in press).

    Kadariya Y., Sementino E., Shrestha U., Gorman G., White J.M., Ross E.A., Clapper M.L., Neamati N., Miller M.S., Testa J.R. Inflammation as a chemoprevention target in asbestos-induced malignant mesothelioma. Carcinogenesis 43:1137-48, 2022. PMID: 36355620 PMCID: PMC10122428.

    Osmanbeyoglu HU, Palmer D, Sagan A, Sementino E, Becich MJ, Testa JR. Isolated BAP1 genomic alteration in malignant pleural mesothelioma predicts distinct immunogenicity with implications for immunotherapeutic response. Cancers (Basel). 16;14:5626, 2022. PMID: 36428720; PMCID: PMC9688367.

    Kurimchak A.M., Herrera-Montávez C., Montserrat-Sangrà S., Araiza-Olivera D., Hu J., Neumann-Domer R., Kuruvilla M., Bellacosa A., Testa J.R., Jin J., Duncan J.S. The drug efflux pump MDR1 promotes intrinsic and acquired resistance to PROTACs in cancer cells. Sci Signal 15:eabn2707, 2022. PMID: 36041010 PMCID: PMC9552188.

    Sementino E., Kadariya Y., Cheung M., Menges C.W., Tan Y., Kukuyan A.M., Shrestha U., Karchugina S., Cai K.Q., Peri S., Duncan J.S., Chernoff J., Testa J.R. Inactivation of p21-activated kinase 2 (Pak2) inhibits the development of Nf2-deficient tumors by restricting downstream hedgehog and Wnt signaling. Mol Cancer Res. 20:699-711, 2022. PMID: 35082167; PMCID: PMC9081258

    Cheung M., Kadariya Y., Sementino E., Hall M.J., Cozzi I., Ascoli V., Ohar J.A., Testa J.R.. Novel LRRK2 mutations and other rare, non-BAP1-related candidate tumor predisposition gene variants in high-risk cancer families with mesothelioma and other tumors. Hum Mol Genet. 30:1750-61, 2021. PMID: 34008015; PMCID: PMC8411985.

    Tan Y., Sementino E., Menges C.W., Kukuyan A.-M., Peri S., Cheung M., Khazak V., Ross E., Fox L.A., Jhanwar S.C., Ramanathan C., Flores R.M., Balachandran S., Testa J.R.  Somatic epigenetic silencing of RIPK3 inactivates necroptosis and contributes to chemoresistance in malignant mesothelioma.  Clin Cancer Res. 27:1200-13, 2021. PMID: 33203643 PMCID: PMC7887036.

    Tan Y., Sementino E., Liu Z., Cai K.Q., Testa J.R.  Wnt signaling mediates oncogenic synergy between Akt and Dlx5 in T-cell lymphomagenesis by enhancing cholesterol synthesis.  Sci Rep. 10:15837, 2020. PMID: 32985581; PMCID: PMC7522078.

    Testa J.R., Berns A. Preclinical models of malignant mesothelioma. Front Oncol. 10:101, 2020. PMID: 32117751; PMCID: PMC7026500.

    Gary JM, Simmons JK, Xu J, Zhang S, Peat TJ, Watson N, Gamache BJ, Zhang K, Kovalchuk

    AL, Michalowski AM, Chen JQ, Thaiwong T, Kiupel M, Gaikwad S, Etienne M, Simpson RM, Dubois W, Testa JR, Mock BA. Hypomorphic mTOR Downregulates CDK6 and Delays Thymic Pre-T LBL Tumorigenesis. Mol Cancer Ther. 19:2221-32, 2020. PMID: 32747423; PMCID: PMC9574474.

    Kukuyan A.-M., Sementino E., Kadariya Y., Menges C.W., Cheung M., Tan Y., Cai K.Q., Slifker M.J., Peri S., Klein-Szanto A.J., Rauscher F.J. III, Testa J.R.  Inactivation of Bap1 cooperates with losses of Nf2 and Cdkn2a to drive the development of pleural malignant mesothelioma in conditional mouse models. Cancer Res. 79:4113-23, 2019. PMID: 31151962; PMCID: PMC6697648.

    Walpole S., Pritchard A.L., Cebulla C.M., Pilarski R., Stautberg M., Davidorf F.H., de la Fouchardière A., Cabaret O., Golmard L., Stoppa-Lyonnet D., Garfield E., Njauw C.N., Cheung M., Turunen J.A., Repo P., Järvinen R.S., van Doorn R., Jager M.J., Luyten G.P.M., Marinkovic M., Chau C., Potrony M., Höiom V., Helgadottir H., Pastorino L., Bruno W., Andreotti V., Dalmasso B., Ciccarese G., Queirolo P., Mastracci L., Wadt K., Kiilgaard J.F., Speicher M.R., van Poppelen N., Kilic E., Al-Jamal R.T., Dianzani I., Betti M., Bergmann C., Santagata S., Dahiya S., Taibjee S., Burke J., Poplawski N., O'Shea S.J., Newton-Bishop J., Adlard J., Adams D.J., Lane A.M., Kim I., Klebe S., Racher H., Harbour J.W., Nickerson M.L., Murali R., Palmer J.M., Howlie M., Symmons J., Hamilton H., Warrier S., Glasson W., Johansson P., Robles-Espinoza C.D., Ossio R., de Klein A., Puig S., Ghiorzo P., Nielsen M., Kivelä T.T., Tsao H., Testa J.R., Gerami P., Stern M.H., Paillerets B.B., Abdel-Rahman M.H., Hayward N.K.  Comprehensive study of the clinical phenotype of germline BAP1 variant-carrying families worldwide. J Natl Cancer Inst. 110:1328-41, 2018.  PMID: 30517737; PMCID: PMC6292796.

     

    Additional Publications

    Open Positions

    About the Position

    The Testa Laboratory at the Fox Chase Cancer Center is seeking a talented and self-motivated Postdoctoral Fellow in Genetics, Cell Signaling, and/or Mouse Models of Cancer. The laboratory uses human mesothelioma specimens and genetically engineered mouse models of mesothelioma to address the following areas: (1) the role of somatic genetic drivers of mesothelioma formation and progression; (2) the impact of cancer-associated genetic alterations on the immune tumor microenvironment and tumor response to targeted  and immunotherapies; (3) the characterization of mechanisms by which BAP1 and other cancer susceptibility genes, e.g., LRRK2, predispose to mesothelioma; and (4) the utilization of preclinical mouse models to evaluate chemoprevention agents, with implications for individuals at high risk of mesothelioma due to hereditary factors and/or exposure to asbestos or other carcinogenic mineral fibers. Some examples of relevant publications from the laboratory are: Testa et al., Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet 2011;43:1022-5; Kadariya et al., Bap1 is a bona fide tumor suppressor; genetic evidence from mouse models carrying heterozygous germline Bap1 mutations. Cancer Res 2016;76:2836-44; Kukuyan et al., Inactivation of Bap1 cooperates with losses of Nf2 and Cdkn2a to drive the development of pleural malignant mesothelioma in conditional mouse models. Cancer Res 2019;794113-23; Cheung et al., Novel LRRK2 mutations and other rare, non-BAP1-related candidate tumor predisposition gene variants in high-risk cancer families with mesothelioma and other tumors. Hum Mol Genet 2021:30:750-61; Kadariya et al., Inflammation as a chemoprevention target in asbestos-induced malignant mesothelioma. Carcinogenesis 2022 Nov 10;bgac089. Additional details regarding the laboratory research can be found at https://www.foxchase.org/joseph-testa.

    The Testa laboratory seeks candidates that have published peer reviewed manuscripts and have strong expertise in molecular biology, cancer biology, genomics and/or mouse modeling.

    About the Training Environment

    As one of the four original cancer centers to receive comprehensive designation from the National Cancer Institute, Fox Chase Cancer Center has been at the forefront of cancer research for almost 90 years. We are home to excellent research facilities, top clinicians and scientists, and outstanding patient care. Our singular focus on cancer, which couples discovery science with state of the art clinical care and population health, remains the foundation of our work.

    The scientist training programs at Fox Chase Cancer Center provide professional development opportunities in four core areas identified as crucial for successful careers in science, research, and health care including communication, leadership, teaching, and mentorship. Upon joining the program, graduate students and postdocs develop individual development plans to help guide their growth. Training throughout the year is supplemented with free professional development opportunities, including a robust ‘How To’ series, writing courses, networking, mentorship, and teaching opportunities, a trainee-led seminar series, a trainee-led annual Research Conference, and more. Postdocs at Fox Chase Cancer Center are supported by the Temple University Postdoc Association and the Office of Academic Affairs at Fox Chase, and are compensated with competitive pay and benefits.

    In addition to the robust training program, scientists at Fox Chase Cancer Center benefit from being part of the rich scientific and biotech environment in the Philadelphia region. Many of our former trainees are now employees (and contacts) at nearby institutions and companies, including The Wistar Institute, Merck, GSK, AACR, and numerous others.

    To Apply

    Email a CV, cover letter that states research interest(s) and goals, and the name of at least three references to [email protected]. Qualified candidates will then be invited to complete a job application.

    The following ratings and reviews are based on verified feedback collected from independently administered patient experience surveys. The ratings and comments submitted by patients reflect their own views and opinions. Patient identities are withheld to ensure confidentiality and privacy. Learn more about our Patient Experience Ratings.

    Ratings Breakdown

    Loading ...

    Patient comments

    Loading ...
    ​​

    This Fox Chase professor participates in the Undergraduate Summer Research Fellowship
    Learn more about Research Volunteering.