March 27, 2024 – Announcements
DF/HCC Prostate SPORE - CEP & DRP RFAs now open! Due May 15, 2024.
CO-LEADER
CO-LEADER
The Dana-Farber/Harvard Cancer Center (DF/HCC) Prostate SPORE seeks to improve the understanding and treatment of prostate cancer. The program consists of three projects, three cores, a career enhancement program (CEP) and a development research program (DRP).
Project 1 leverages tumor specimens from patients with high-risk prostate cancer treated with neoadjuvant therapies to understand how tumors respond and resist acute potent androgen receptor blockade and to develop novel strategies to improve cure rates and combat resistance.
Localized high-risk prostate cancer (PCa) comprises ~15% of newly diagnosed localized PCa, and the cure rate for these tumors after primary surgical treatment by radical prostatectomy (RP) is suboptimal, with recurrent disease in up to ~50%. Neoadjuvant trials for these localized high-risk PCa may provide valuable opportunities to increase cure rates and our understanding of response and resistance. However, previous studies of neoadjuvant androgen deprivation using GnRH agonists alone have generally shown infrequent pathological complete responses (pCRs) and have not shown evidence of clinical efficacy. We have hypothesized that responses to neoadjuvant ADT have been limited due to substantial residual androgen in the prostate, and that more intensive neoadjuvant androgen signaling inhibition (ASI) therapy, in addition to increasing pCR rates, will translate into an improvement in disease free survival (DFS) and ultimately overall survival (OS).
We have been testing this hypothesis in a series of phase 2 trials examining the efficacy of neoadjuvant intensive ASI using leuprolide in combination with abiraterone (ABI) and/or enzalutamide (ENZ) or apalutamide (APA) for 6 months prior to radical prostatectomy (RP) in men with localized high-risk localized PCa. Responses appear to be improved relative to previous neoadjuvant trials, with pCR rates of 10-12%, and with an additional 10-20% having minimal residual disease (MRD). Moreover, compared to expected relapse rates, patients with pCR/MRD have had much lower than predicted rates of PSA failure and progression to metastatic PCa. However, it remains unclear whether the intensive ASI therapies are preventing the emergence of metastatic disease (presumably by treating microscopic metastatic disease) versus the possibility that the cases with pCR/MRD reflect tumors that have less metastatic potential. Moreover, the molecular basis for residual disease in prostate, and its relationship to metastatic disease in patients who progress, remain to be determined.
To address the genomic basis of response versus resistance, we propose comprehensive genomic analyses of tumors with exceptional responses (pCR/MRD) to intensive neoadjuvant ASI therapy, and of residual disease in RP specimens in men who do not achieve pCR/MRD, including determining whether oncogenic alterations in the residual disease are driving the subsequent emergence of metastatic PCa (Aim 1). These studies will leverage samples from our previous trials, as well as a large multicenter phase 3 study of neoadjuvant GnRH agonist/antagonist combined with APA in comparison with GnRH agonist/antagonist alone (PROTEUS trial, supported by Janssen, NCT03767244). Aim 2 will identify actionable acute nongenomic adaptations that mediate initial resistance to intensive ASI therapy, and specifically test the hypothesis that these adaptations converge on expression of D cyclins and activation of CDK4/6 to drive proliferation. Aim 3 is a randomized phase 2 trial of intensive ASI (leuprolide plus darolutamide, DARO) alone or in combination with the CDK4/6 inhibitor abemaciclib.
Our long-term goal is to establish neoadjuvant trials as a platform for assessing the efficacy of novel combination therapies in castration-sensitive PCa. Bringing effective systemic therapies to the neoadjuvant setting has the potential to increase cure rates for men with localized high-risk disease, who may be rendered disease free by the therapy or who may benefit from subsequent adjuvant therapy guided by findings in the RP. Moreover, validating that pathological responses after neoadjuvant therapy are predictive of DFS and ultimately OS has the potential to markedly accelerate testing of novel agents and combinations through neoadjuvant trials, which may then also be effective in metastatic castration-sensitive PCa. Finally, insights gained into tumor biology and clonal dynamics can be exploited to develop new therapeutic strategies. The Specific Aims and subaims are as follows:
Aim 1. Identify molecular features of tumors that correlate with pathological responses to neoadjuvant intensive androgen signaling inhibition (ASI) and its clinical significance.
a. Determine molecular features of tumors that are associated with pathological complete responses.
b. Identify features predictive of improved disease free survival.
c. Identify genomic mechanisms of resistance to neoadjuvant intensive androgen signaling inhibition and their contribution to metastatic disease.
Aim 2. Identify actionable nongenomic mechanisms of resistance to neoadjuvant intensive androgen signaling inhibition.
a. Identify acute adaptations to intensive androgen signaling inhibition.
b. Assess efficacy and mechanism of action of CDK4/6 inhibitors combined with castration in PDX models.
Aim 3. Assess the efficacy of neoadjuvant androgen signaling inhibition combined with abemaciclib.
a. Assess efficacy of neoadjuvant intensive ASI therapy combined with abemaciclib.
b. Assess target engagement and mechanisms of intrinsic or acquired resistance.
Project 2 will develop innovative strategies to target the epigenome in later stages of advanced castration resistant prostate cancer and will develop a first-in-field clinical trial focused on co-targeting EZH2 and PARP.
Despite significant advances in prostate cancer therapy, available treatment options particularly in later stages of the disease, are still limited and the treatment-resistant setting represents a serious unmet medical need. Resistance to androgen receptor (AR) therapies in prostate cancer is predominantly AR-driven. Non-AR driven castration resistant prostate cancer (CRPC) represents approximately 15-20% of advanced disease and is more predominant after potent AR-targeted therapies; one emerging mechanism is through lineage plasticity with reprogramming of prostate adenocarcinoma tumor cells towards a neuroendocrine phenotype. Epigenetic dysregulation, including changes in DNA methylation and overexpression of the enhancer of zeste homolog 2 (EZH2) also drive castration resistance, though much remains to be understood regarding the impact and contextual timing of these epigenetic changes in mediating AR-driven versus non-AR-driven resistance. EZH2 is a member of a polycomb repressor complex 2 that plays an important role in development through transcriptional repression via histone methylation. EZH2 is overexpressed in CRPC, both in adenocarcinoma and neuroendocrine prostate cancer (NEPC), along with EZH2 activity measured by histone H3 lysine 27 trimethylation. Beyond its canonical role, we previously identified a non-canonical role of EZH2 in activating AR signaling (mediated in part by phosphorylation) as well as in driving lineage plasticity and NEPC (in part through cooperation with N-myc). Importantly, EZH2 is targetable and EZH2 inhibitors are currently in clinical trials. The EZH2 inhibitor tazemetostat has shown clinical activity in non-Hodgkin lymphoma, synovial sarcoma, mesothelioma, and was recently FDA approved for the treatment of epithelioid sarcoma. The prostate cancer patient subset most likely to benefit from EZH2 inhibition has not been established. Current data suggests that single agent activity may be limited and that combination strategies should be pursued. We recently discovered a novel interaction between EZH2 and DNA repair processes and a synergy between EZH2 and PARP inhibition in CRPC. We hypothesize that overexpression of EZH2 during CRPC progression drives downstream molecular programs through both canonical and non-canonical mechanisms, and that these downstream effects are context dependent. We hypothesize that combination therapy with EZH2 and PARP inhibition will have differential biologic impact in CRPC based on the underlying genomic, epigenomic, and phenotypic context.
Aim 1: Elucidate the crosstalk between EZH2 and DNA repair pathways and mechanisms of synthetic lethality. We found that DNA repair genes are induced by EZH2 and acutely repressed by EZH2i, and the combination of EZH2 with PARP inhibition is synergistic. We will examine the canonical regulation by EZH2 of DNA repair pathways (NHEJ, HR) and the effects of EZH2 inhibition on de-repression of MAD2L2 and SLFN11 as a means to sensitize to PARP inhibition. We will also investigate non-canonical regulation of DNA repair genes through EZH2 interaction with cofactors such as E2F and AR. We will further evaluate response to EZH2i and EZH2i+ PARPi and downstream effects in homologous recombination (HR) intact and HR-deficient patient derived xenograft models. Results from this Aim will provide a novel molecular route for PARPi synthetic lethality in HR proficient prostate cancer.
Aim 2: Define the biologic impact of EZH2 + PARP inhibition across the heterogeneous spectrum of CRPC including both AR-driven and non-AR-driven tumors. We have observed synergy when co-targeting EZH2 and PARP1 in AR-driven CRPC models. We will extend this combination to models that represent the CRPC spectrum, including NEPC. We will assess consequences of EZH2i and effects of combination therapy on tumor growth, DNA damage, downstream gene expression profiles, DNA methylation, and chromatin accessibility. We will specifically evaluate EZH2 targets and PRC dependence, changes in AR and neuroendocrine programs, as well as heterogeneity at the single cell level. Results from this Aim will provide insights into the downstream consequences of EZH2i and PARPi therapy across distinct subsets of CRPC.
Aim 3: Identify biomarkers of response and resistance to the combination of EZH2 + PARP inhibition in patients. We will conduct investigator-initiated Phase 1/2 clinical trials of the combination of the EZH2i tazemetostat with the PARPi talazoparib to assess the safety, efficacy, and biomarkers of response in patients with CRPC/NEPC. Pre-treatment and on-therapy biopsies will be assessed for changes in g-H2AX on therapy, as a primary pharmacodynamic marker supporting mechanism. Baseline tumors will also be classified based on their underlying genomic (DNA repair/AR/RB) and epigenetic (NE/adeno) status and correlated with response and duration of therapy. We will evaluate other specific DNA damage readouts (eg., RAD51, SLFN11, MAD2L2, phospho-RPA) and downstream changes in signaling/expression profiles (eg., NHEJ/HR repair genes, PRC targets, AR signaling, NE programs) on therapy. Results from this Aim will provide first-in-field clinical data of combined EZH2 and PARP inhibitor therapy in patients with advanced prostate cancer.
Project 3 delves deep into biomarkers in localized prostate cancer, leveraging innovations in computation and biologically-guided deep learning, to deliver on precision cancer medicine in this disease state. The ability to understand why some localized prostate cancers are phenotypically aggressive and predict which localized prostate cancers will behave in this manner addresses a large clinical unmet need.
Over 90% of the 268,490 estimated cases of prostate cancer (PCa) in 2022 will present as localized disease, with a majority defined as intermediate or high-risk for recurrence based on traditional pathologic features1-4. Despite standard of care therapy commonly involving radical prostatectomy (RP) or combined radiation and hormonal therapy, the risk of recurrent disease is as high as 50% for subsets of men with localized, high-risk PCa, and more than half of PCa deaths occur in men with initially localized or locally advanced disease. In the preceding decade, multiple molecular determinants of lethal prostate cancer have been identified, including germline and somatic mutations in DNA repair genes that impact genome-wide and immune microenvironmental molecular signatures in retrospective cohorts5-14. Understanding the biological relevance of these properties and their co-occurring interactions, and as well as their relationship to existing tissue-based and clinical biomarkers for determining at the point-of-care which patients are likely to progress to metastasis and death despite typical management, is essential. Similarly, the extent to which these features imprint spatial patterns in PCa histopathology, and whether prognostic patterns can be learned from these data for clinical use, is uncertain but may dramatically impact worldwide practice patterns given the ubiquity of histopathology. Broadly, two major activities would significantly advance localized PCa care: (i) understanding the convergent biological underpinnings behind phenotypically aggressive but clinically localized PCa to inform new therapeutic paradigms, and (ii) predicting which localized PCa will behave in this manner to enrich for adjuvant studies geared towards improving survival for those at highest risk of progression to lethal disease. Taken together, tackling these challenges would significantly advance basic prostate cancer research and impact the development of personalized escalation and de-escalation treatment strategies for patients. Innovations in explainable artificial intelligence, paired with the advent of harmonized and representative clinicogenomic PCa cohorts, has created a new opportunity to engage in hypothesis-driven reframing of localized PCa. Our overarching hypothesis is that interacting molecular and histopathologic features determine prognostic outcomes in intermediate and high-risk localized PCa after RP, and that biologically guided deep learning models of molecular, pathologic, and phenotypic data can distinguish these clinical states. Our interdisciplinary team will leverage diverse cohorts to address this hypothesis through these Specific Aims:
Aim 1: Define the interacting germline and somatic properties and co-occurring alterations that mediate aggressive localized prostate cancer using biologically guided neural networks
Hypothesis: Co-occurring known and novel germline and somatic alterations discriminate aggressive localized primary PCa after RP, and these patterns can be revealed through biologically guided neural networks.
1A: Identify germline mutation patterns in DNA repair and other canonical PCa pathways in large and ancestrally diverse prostate cancer cohorts using deep learning models
1B: Determine the co-occurring molecular properties that mediate metastasis and overall survival (MFS/OS) outcomes using biologically guided interpretable deep learning
1C: Functionally validate candidate co-mutation events identified through molecular deep learning.
Aim 2: Determine the histopathologic latent representations of molecularly and clinical distinct localized prostate cancer through digital pathology deep learning
Hypothesis: The spatial histopathologic architecture of intermediate/high risk localized PCa and its surrounding microenvironment reflect specific underlying molecular states, and deep learning models of histopathology images can predict these molecular states and learn graphical patterns that discriminate MFS/OS outcomes.
2A: Determine the tumor and microenvironmental spatial characteristics of distinct PCa molecular subtypes
2B: Develop computer vision models that predict MFS/OS in localized PCa after RP from histopathology images of RP specimens
2C: Implement histopathology-based deep learning predictive models in PCa pathologist clinical settings
Aim 3: Evaluate condensed deep learning genomic models for predicting MFS/OS in retrospective and clinical trial prostate cancer cohorts
Hypothesis: Deep learning genomic models developed with clinically validated and widely available NGS panels can stratify MFS/OS outcomes in intermediate and high risk localized PCa after RP beyond of established histopathologic and clinical prognostic risk factors.
3A: Compare the prognostic performance of a condensed P-NET model with existing histopathologic and clinical models in retrospective real-world cohorts
3B: Evaluate the prognostic performance of a condensed P-NET PCa model in the Phase 3 CALGB 90203 trial Successful execution of this proposal will transform our biological understanding of PCa and our ability to develop future surveillance and adjuvant strategies for men at highest risk of metastatic relapse and death from PCa.
Broadly, this project will transform precision cancer medicine for localized PCa and serve as a model for the creation, development, and application of these emerging methodologies across the disease continuum.
The DF/HCC Prostate SPORE Administrative Core (Core A) is instrumental for the overall function and success of the Prostate SPORE program. The Directors will work closely with Advisory Board Members, Patient Advocates, and SPORE investigators to foster an environment that promotes collaboration and transparency in research. Regular reviews and fiscal oversight by Core A will ensure that the program has the resources it needs to be successful. Core A will also ensure that the SPORE integrates well with the broader DF/HCC program and is capable of leveraging resources and institutional infrastructure to accelerate translational prostate cancer research.
Dana-Farber Cancer Institute
Beth Israel Deaconess Medical Center
Administrative Support Specialist
The goal of the Administrative Core is to ensure the overall success of the DF/HCC Prostate SPORE in achieving its mission to catalyze and accelerate impactful translational research that impacts the treatment and outcomes for individuals with prostate cancer.
Aim 1: Provide necessary resources and fiscal oversight. Core A will be responsible for the overall management and administration of the SPORE under the leadership of SPORE Co-Directors Drs. Beltran and Balk, SPORE investigator Dr. Timothy Rebbeck, and lead patient advocate Mr. Thomas Farrington. The Core will be supported by an administrator to help with the planning and organization of SPORE translational research meetings and other events, coordination of IAB and EAB and executive meetings, patient advocacy meetings, communication, and other goals of Core A. There will also be a dedicated grant administrator at DFCI who will ensure that standard administrative procedures will be applied for the overall management, planning, monitoring, fiscal oversight, and reporting of the SPORE activities. The Core will oversee the management of key personnel, monitor progress as well as interactions between the projects and cores, and guide their development. Core A will work with DRP and CEP programs to distribute RFAs, organize review panels, allocate funding, and ensure diversity of applicants and mentors.
Aim 2: Monitor research progress and plan for the future. Research progress will be reviewed regularly by the Administrative Core to ensure projects are proceeding according to their goals, and if not, that changes are made (i.e., addition of resources, modification of research focus) to produce successful results. Reviews will also ensure proper regulatory compliance, patient consenting and data sharing. Research updates will be formally presented annually to the Internal Advisory Board and External Advisory Board. In addition, ongoing evaluation of research progress will continue through presentations at the monthly SPORE Research-in-Progress meetings, the annual DF/HCC prostate cancer retreat, and during scheduled meetings between project/core leaders, SPORE leadership, and patient advocates. The Administrative Core will organize these meetings and maintain reports. Additional forums and outreach related to women in prostate cancer research, underrepresented ethnic and racial groups, and the community will also be supported to ensure diverse representation of both researchers and patients. Core A will also be the primary point of contact with NCI, providing a flow of information regarding progress and scientific advances.
Aim 3: Integrate the SPORE within the Dana Farber/Harvard Cancer Center. By providing access to DF/HCC shared resources, clinical trials infrastructure, and educational and outreach initiatives, as well as providing dedicated institutional funding to the SPORE, DF/HCC will help catalyze research and accelerate progress within the Prostate SPORE program. In turn, the Prostate SPORE will also provide added value to the DF/HCC community at large by providing core resources, funding for pilot projects, and fostering collaborations and career development. Core A will work closely with DF/HCC to link the SPORE website and social media pages and to announce educational symposia, research meetings, new RFAs, and patient educational meetings, and other SPORE activities throughout the DF/HCC community and the greater Boston area.
Aim 4: Foster collaborative research and dissemination of research findings. The Administrative Core will help disseminate research, facilitate data sharing, and foster collaborations within the SPORE, DF/HCC at large, and with other SPORE programs. The core will support monthly translational research meetings, an annual prostate cancer retreat, a women in science forum, inter-SPORE meetings, and other events. The core will nominate SPORE investigators for presentations, support attendance and participation, and encourage women and underrepresented researchers to participate. We hope these interactions will encourage investigators from diverse backgrounds to collaborate, establish multidisciplinary mentorship for young investigators, and continue to foster a culture of diversity and translational research.
Aim 5: Integrate a patient perspective throughout all research activities in the Prostate SPORE. The SPORE is committed to patients and will work to ensure the patient perspective is incorporated throughout all research activities in the SPORE. To this end, we have assembled a Prostate Cancer Patient Advocacy Committee. We currently have seven members on the Committee and will continue to grow this program as part of the SPORE. SPORE patient advocates will be integrated and provide guidance to the SPORE in all aspects of the program. Enrollment of a diverse patient population, including underrepresented racial and ethnic groups, in SPORE-related trials is a major priority.
The Biostatistics and Computational Core (Core B) of the Dana-Farber/Harvard Cancer Center (DF/HCC) Prostate SPORE interacts with all research activities within the SPORE, including the SPORE Projects, Developmental Research Program, and Career Enhancement Program to ensure the highest standards of scientific rigor in areas of study design, data management and integrity, and data analysis and interpretation.
Dana-Farber Cancer Institute
Dana-Farber Cancer Institute
The Dana-Farber/Harvard Cancer Center (DF/HCC) Prostate SPORE Biostatistics and Computational Biology Core collaborates and provides consultation on all research activities within the SPORE including SPORE Projects, the Developmental Research and Career Enhancement Programs, and other SPORE Cores to ensure the highest standards of scientific rigor in areas of study design, data management and integrity, and data analysis and interpretation.
The specific aims are to:
Organizing biostatistical and computational biology expertise as a shared resource core is a cost-effective approach to ensure that collaboration is readily available to SPORE investigators and is an effective strategy to guarantee a high degree of integration among projects, which have interrelated analytic goals and needs.
Acquisition and characterization of biospecimens linked with clinical data and the development of patient- relevant preclinical models, are essential components for effective and impactful translational research. The Dana Farber/ Harvard Cancer Center (DF/HCC) Prostate SPORE Biospecimen Core (Core C) will maintain and expand a mature prostate cancer biorepository, including blood and tissue specimens, from a diverse group of prostate cancer patients across DF/HCC who have provided consent to our clinical database and biobank protocols. Core C will provide tissue specimens and vast pathology expertise to SPORE investigators including the evaluation of both human and mouse model tissues.
The first and foremost goal of the DF/HCC Prostate SPORE Biospecimen Core is to maintain and expand the existing repository for specimens, including blood and tissue, from a diverse group of prostate cancer patients who have provided consent to our clinical database and biobank protocols. Included in this component are the collection, freezing, and storage of fresh prostate cancer specimens and paired non tumor tissue; the collection, processing, and storage of blood; the identification and provision of samples of fixed tissues, including construction of tissue microarrays (TMAs), from biopsy and prostatectomy samples obtained from patients who have consented to allow analysis of these tissues. The caTissue system, which is the NCI caBIG's biorepository tool for biospecimen inventory management, is currently used to track specimens through every step of the requesting, shipping, and receiving process through the use of barcode technology.
Importantly, the Core will continue to maintain a database of clinical data (CRIS) on all consented patients with prostate cancer. The value of the database is enhanced by the use of standardized pathology review procedures and data collection procedures. The database and specimen tracking system allow seamless sharing of specimen resources, de-identified and linked to clinical outcome data, behind a secure data management system that is available to SPORE investigators at all participating institutions - namely Dana-Farber/Harvard Cancer Center (DF/HCC) hospitals, including the Dana-Farber Cancer Institute and Brigham and Women's Hospital (DFCI/BWH), the Beth Israel Deaconess Medical Center (BIDMC), Harvard School of Public Health (HSPH), and Massachusetts General Hospital (MGH). The protection of patient confidentiality is guarded throughout the whole process, from specimen collection to use in research projects. The Biostatistics/Computational Biology Core is and will continue to be responsible for assisting in the data analysis, data auditing and quality control.
Finally, the Core will provide SPORE investigators a variety of services critical to successful molecular analysis of human prostate tumors as well as animal model tissues. These services include: histopathologic review and quality control analysis of all tumor samples utilized in experimental studies; macrodissection of frozen tissue blocks and slide macrodissection of formalin-fixed paraffin-embedded (FFPE) or frozen tissues to ensure high neoplastic cellularity for samples utilized in experimental studies; laser capture microdissection {LCM) to provide ultra-pure tumor samples; extraction of DNA/RNA from both FFPE and frozen tissues; performance of immunohistochemistry {IHC) and multiparametric immunofluorescence {IF) stains on human prostate cancers (TMAs or whole tissue sections); optimization and validation of antibodies to known and novel proteins for use in IHC and IF; analysis of a broad range of IHC and IF stains using computer-assisted image analysis.
The Biospecimen Core thus has three specific aims:
Specific Aim 1: To provide a specimen bank for SPORE investigators
Specific Aim 2: To maintain a database of key clinical variables
Specific Aim 3: To provide pathology services to SPORE investigators
The career enhancement of early-career faculty is essential for continued progress and success of the Dana- Farber/Harvard Cancer Center (DF/HCC) Prostate SPORE. Supporting and mentoring the next generation of leaders in basic, clinical, population, and translational prostate cancer research is a top priority for the SPORE. The Career Enhancement Program (CEP) will provide a formalized mechanism to support this mission. Through its activities, the CEP will continue to develop a pipeline of promising talent within DF/HCC, select innovative projects for funding, assist in building collaborations to facilitate clinical translation, provide formalized mentorship to enhance the career development of early career investigators, and provide opportunities to sustain the careers of these individuals in translational prostate cancer while retaining them within DF/HCC. The CEP will also identify investigators who research other cancers seeking to transition into a career in prostate cancer. A key CEP goal is to attract, retain, and nurture young talented researchers with the greatest potential for innovation and sustained contributions to the field, with an emphasis of supporting diversity.
The CEP is led by Drs. Paul Nguyen (BWH/DFCI), Lorelei Mucci (HSPH), and Adam Feldman (MGH), established prostate cancer investigators who are passionate about mentoring and have successfully mentored numerous trainees and faculty. They have established a formal program for the SPORE CEP to broadly solicit proposals, and target investigators including women and under-represented racial and ethnic groups (UREG). They have established a UREG Advisory Committee that will formally review the CEP Program, give input on mentorship activities, and provide expert guidance on diversity initiatives including strategies for outreach to diverse candidates outside of DF/HCC. In addition, the CEP will implement initiatives to broadly support and mentor early career women in prostate cancer, an effort led by Drs. Mucci and Beltran. CEP proposals will undergo a fair and transparent review. Awardees will benefit from research funding, access to SPORE infrastructure and cores, exposure and interactions with prostate cancer investigators through SPORE meetings, educational sessions, and through mentoring activities. The SPORE as whole will also benefit from interactions with diverse talent and fresh ideas. We expect that the CEP program will play a central role in the DF/HCC Prostate SPORE and will foster the next generation of leaders in prostate cancer translational research.
*Click the Read More button below to see publications
*Coming Soon
March 27, 2024 – Announcements
DF/HCC Prostate SPORE - CEP & DRP RFAs now open! Due May 15, 2024.
January 02, 2024 – Prostate Cancer
The goal of the DF/HCC Prostate SPORE Developmental Research Program (DRP) is to identify and support innovative and exciting prostate cancer research, in order to maximize the chance that new and cutting-edge concepts supported by promising pilot data have the capability to ultimately impact the field. Support from the DRP will allow projects to develop the foundations needed for mature, hypothesis-driven proposals with high translational potential that will be competitive for additional funding as future full SPORE projects and/or national peer reviewed grants. The DRP will also create opportunities for junior faculty or senior investigators from other fields to transition into prostate cancer research.
The DRP will work in concert with the Career Enhancement Program (CEP) to support emerging and established investigators from underrepresented racial and ethnic groups as well as projects that address the disparate prostate cancer outcomes of patients seen in these groups. The objective is to stimulate interest from diverse disciplines and backgrounds to bring fresh ideas and new approaches to prostate cancer. As such, researchers from diverse backgrounds, including women and investigators from underrepresented racial and ethnic groups, will be encouraged to apply. DRP Award recipients will receive two years of funding as well as access to SPORE resources. A standing committee made up of diverse disciplines and backgrounds, highly qualified, and engaged SPORE and DF/HCC investigators will solicit projects broadly, review applications, select meritorious awards, and review progress. DRP recipients will become part of the SPORE community with continued active engagement, feedback, and exposure.
*Coming Soon
March 27, 2024 – Announcements
DF/HCC Prostate SPORE - CEP & DRP RFAs now open! Due May 15, 2024.
January 02, 2024 – Prostate Cancer