Skip to Content
Translational Research Program (TRP)
Contact CIP
Show menu
Search this site
Last Updated: 10/10/18

Moffitt Skin Cancer SPORE

H. Lee Moffitt Cancer Center and Research Institute

Principal Investigator: Vernon Sondak, M.D.

Principal Investigator Contact Information

Vernon Sondak, M.D.
Chair, Department of Cutaneous Oncology
12902 Magnolia Drive
H. Lee Moffitt Cancer Center & Research Institute
Tampa, FL 33612
Phone: 813-745-8788
Fax: 813-745-7211
Email: vernon.sondak@moffitt.org

OVERALL

Extraordinary progress has been made in the last decade in understanding the biology of melanoma and other cutaneous malignancies, and as a consequence, in developing new therapies for them. In the last few years, three new drugs were approved for metastatic or resected melanoma (ipilimumab, sylatron and vemurafenib), all based on strong pre-clinical proof of concept data that led ultimately to definitive pivotal trials. Regardless of those impressive achievements, the outlook for, and median survival of, patients with metastatic melanoma and recurrent non-melanoma skin cancer remain poor. The use of genetic analysis to choose melanoma patients for BRAF inhibitor and other targeted treatments is becoming increasingly widespread, but the large proportion of melanoma patients that are BRAF wild-type, or that are BRAF mutated and relapse after initial therapeutic success, and the very large number of non-melanoma skin cancer patients in this country indicate that new approaches to the therapy of cutaneous malignancies are sorely needed. Therefore, the Moffitt Cancer Center Skin SPORE will 1) carry out 3 SPORE projects, each of which is based around an innovative, fundamental and scientifically exciting observation made by our own researchers and ready to be translated into phase I or II trials; 2) utilize patient specimens from those trials to iteratively improve our approach to cutaneous malignancies, and 3) prospectively assess the clinical utility of these approaches, each of which has the potential to impact the way we approach therapeutic and prevention interventions for metastatic melanoma or non-melanoma skin cancers.

The Moffitt Skin SPORE has three translational research projects that have a common thematic approach designed to promote progress in the prevention and treatment of cutaneous malignancies based on promising developments in our laboratories that will be applied to clinical trials in this SPORE that are likely to have an impact in the field of cutaneous malignancies. Our overarching goal in this Skin SPORE is to develop ways to potentiate promising targeted and immunologic therapies for cutaneous malignancies. In the three melanoma projects, fundamental observations made at Moffitt Cancer Center about how activated cell therapy with tumor infiltrating lymphocytes (Project 1), ipilimumab (Project 3) and BRAF inhibitors (Project 2) are being explored in both pre-clinical models and in clinical trials for patients with metastatic disease. Cutting edge pre-clinical models including innovative in vitro and animal models will be used to optimize these strategies. Biospecimens from patients on ongoing and planned trials already testing these concepts will enhance our ability to go from bench to bedside and back. The projects are supported by three cores; Tissue, Pathology and Bioinformatics Core, Biostatistics Core and Administration/Clinical Trials Core. The Moffitt Skin SPORE also has a Developmental Research Program to support pilot projects that exploit new research opportunities in cutaneous malignancies for established cutaneous investigators and non-cutaneous scientists new to the field, and a Career Development Program to promote and support the cutaneous malignancy research efforts of junior faculty and established investigators who will re-focus their research.

PROJECT 1: Potentiating the effects of targeted and cytotoxic agents on cell-based immunotherapy in melanoma

Project Co-Leaders and Co-Investigators:
Dmitry Gabrilovich, M.D., Ph.D.
Jeffrey Weber, M.D., Ph.D.

Adoptive cell therapy with TIL is a promising therapy for melanoma, but is practical only for a limited cohort of patients with stage IV disease. In this project we propose preclinical experiments and two clinical trials to increase the efficacy of this treatment and decrease the likelihood that patients will drop out prior to receiving their TIL, increasing the applicability of this technology to patients with stage IV melanoma. The tumor microenvironment creates powerful obstacles for cancer immunotherapy by limiting the expansion of local tumor-specific T lymphocytes, preventing T-cell penetration and the killing of tumor-specific targets. Therefore, therapeutic targeting of the tumor microenvironment combined with immunotherapy is a logical and attractive approach. We have shown that both chemotherapy and treatment targeted to mutated BRAF increased the susceptibility of melanoma cells to the cytotoxic effect of T cells through a dramatic perforin-independent increase in permeability to granzyme B (GrzB) released by activated cytotoxic T cells (CTL). Our data strongly suggested that this effect was mediated via up-regulation of the expression of mannose-6-phosphate receptors (MPR) on the surface of tumor cells while undergoing autophagy. When combined with chemo- or targeted therapy, CTLs raised against specific antigens were able to induce apoptosis in neighboring tumor cells that did not express those antigens, creating a bystander effect. Our preliminary experiments demonstrated that BRAF inhibitors with potent anti-tumor activity against melanoma induced up-regulation of MPR in BRAF mutated melanoma cells, suggesting that they might provide a potent bystander effect with adoptive cell therapy. In the current proposal we will further explore the mechanisms by which targeted and chemotherapeutic agents alter tumor susceptibility to T cell destruction and propose two clinical trials. In the first trial we will biopsy accessible tumors from patients receiving either a targeted BRAF agent or a chemotherapeutic drug before and after treatment and determine if MPR, markers of autophagy and GrzB are increased on melanoma cells, and when after initiation of treatment increases in these markers can be measured. These findings will then be translated to a phase II trial of the BRAF inhibitor vemurafenib combined with adoptive cell therapy with tumor infiltrating lymphocytes (TIL) and IL-2 after lymphdepletion. This trial will test whether targeted therapy combined with TIL will increase the clinical efficacy of the immunotherapy and result in a higher proportion of patients who do not drop out before they receive their TIL.

Specific Aims:

Three specific aims are proposed to test this hypothesis. Specific Aim 1 will identify the cellular and molecular mechanisms by which the expression of MPR on melanoma cells is induced by BRAF inhibition and chemotherapy and to establish its importance for the efficacy of targeted- or chemo-immunotherapy. Specific Aim 2 will study the effect of targeted therapy with BRAF inhibitors or chemotherapy with taxanes on regulation of MPR and autophagy on tumor cells in patients with melanoma. Finally, Specific Aim 3 will determine if tumor MPR, autophagy and granzyme B expression are associated with response to combined adoptive T-cell therapy and BRAF inhibition in patients with melanoma. This project relies on the use of Tissue, Pathology and Bioinformatics Core A (evaluation of MPR and GrzB expression and autophagy in tumor tissues isolated from patients treated with a BRAF inhibitor or conventional chemotherapy), Administrative/Clinical Trials Core C (clinical trial of adoptive T-cell therapy and BRAF inhibitor vemurafenib) and Biostatistics Core B (statistical analysis of animal experiments and results of clinical trials).

PROJECT 2: Abrogation of therapeutic escape pathways in BRAF mutant melanoma

Project Co-Leaders and Co-Investigators:
Keiran Smalley, Ph.D.
Vernon Sondak, M.D., Ph.D.
David Basanta, Ph.D.
John Koomen, Ph.D.
Rix Uwe, Ph.D.

Although small molecule inhibitors of BRAF and MEK and the BRAF+MEK inhibitor combination are showing great promise as novel melanoma therapies, their effectiveness is severely limited by both acquired and intrinsic drug resistance. The aim of the proposal is to use phospho-proteomics, chemical proteomics and network modeling to identify the key mechanisms of therapeutic escape and to perform preclinical experiments and to analyze specimens from an ongoing phase I clinical trial to determine whether inhibition of HSP90 abrogates the onset of BRAF inhibitor resistance.

Our long-term goal is to develop therapeutic strategies that improve the survival of patients with disseminated melanoma by potentiating new and existing targeted therapies. Despite the impressive responses achieved in BRAF mutant melanoma patients treated with BRAF inhibitors, resistance inevitably occurs. A number of potential resistance mechanisms have been described, the majority of which bypass mutant BRAF through the reactivation of MAPK and PI3K/AKT signaling. Although current clinical strategies are focused upon the use of BRAF inhibitors in conjunction with MEK or PI3K inhibitors, the development of multiple, subtle signaling alterations at many nodes within the melanoma signaling network is likely to result in eventual resistance even to these combinations. Conceptually, we believe that the adaptive signals that mediate resistance in the majority of cases are both “tumor-intrinsic”, resulting from the rewiring of the melanoma signal transduction network, as well as “host-derived”, mediated by altered growth factor secretion and extracellular matrix from stromal fibroblasts. Our working hypothesis is that long-term abrogation of resistance will only be achieved if BRAF can be targeted in addition to multiple receptor tyrosine kinase (RTKs) and cell/matrix adhesion signals. Our preliminary studies suggest that most if not all of the signaling proteins implicated thus far in the escape from BRAF inhibitor therapy are clients of heat shock protein (HSP)-90, and we showed that inhibition of HSP90 was effective at preventing and overcoming resistance both in vitro and in vivo. In this proposal, we will use innovative phosphoproteomic- and chemical proteomic-based systems biology approaches to define the HSP “clientome” that drives intrinsic and acquired resistance of melanomas to MAPK pathway inhibition (BRAF, MEK and BRAF+MEK). We will further investigate the role of fibroblast-derived signals in remodeling the HSP-clientome of melanoma cells and will determine how this allows the tumor to escape MAPK inhibitor-mediated apoptosis. Pre- and post-treatment biopsies from melanoma patients receiving a BRAF and HSP90 inhibitor combination (vemurafenib+XL888) will be analyzed to look for patterns of HSP client protein degradation associated with long-term therapeutic response. Together, these studies are expected to give a systems level view of how melanoma cells resist MAPK pathway inhibition and will provide new paradigms to overcome drug resistance in melanoma.

Specific Aims:

The goal of Specific Aim 1 is to use comprehensive proteomic and chemical proteomic profiling to identify the key HSP90 client proteins required for the escape from MAPK pathway inhibitor therapy. In Specific Aim 2 we will determine the role of HSP90 client proteins in microenvironment-mediated drug resistance in melanoma. Finally, in Specific Aim 3 we will determine the pattern of HSP90 client degradation and examine its association with response in patient specimens from the phase I trial of vemurafenib in combination with the HSP90 inhibitor XL888.

PROJECT 3: Augmenting the immunogenicity of melanoma through manipulation of histone deacetylases (HDACs)

Project Co-Leaders:
Eduardo Sotomayor, Ph.D., M.D.
Ed Seto, Ph.D.
Jeffrey Weber, M.D., Ph.D.

In 2012, metastatic melanoma remains largely incurable. As such, innovative therapies based in a better mechanistic understanding of melanoma immunobiology are greatly needed. We strongly believe that our project would lead to novel epigenetic-based approaches to overcome T-cell tolerance and potentiate further the efficacy of CTLA4 blockade in patients with metastatic melanoma.

In spite of the progress made in the understanding of the biology, genetics and immunology of melanoma, the outcome for patients with advanced-stage disease has remained poor. A step forward toward better therapies was recently provided by the improvement in overall survival observed in melanoma patients treated with an anti-CTLA4 antibody. Attempts to further augment the efficacy of this treatment would still face however a variety of immunosuppressive factors operative in melanoma-bearing hosts. Among those, one that has gained much attention is the ability of melanoma tumors to induce T-cell tolerance. Our studies to date of the epigenetic regulation of T-cell unresponsiveness point to histone deacetylase inhibitors (HDI) as promising immunomodulatory compounds given their dual ability to influence the immunogenicity of melanoma tumors and enhance T-cell function. These observations together with our additional findings that HDAC6 and HDAC11 regulates melanoma immunogenicity and T-cell responsiveness respectively, provided the rationale to mechanistically address the role of HDACs in melanoma immunobiology. The hypothesis to be tested is therefore whether epigenetic manipulation of specific HDACs might augment the immunogenicity of melanoma cells and/or augment T-cell responses leading to breaking of immune tolerance and enhancement of the efficacy of CTLA4 blockade. The animal models, molecular and pharmacological tools we have in hands together with the access (through the Pathology Core of this SPORE) to human melanoma samples would allow us to gain insights into the role of HDAC6 in melanoma proliferation, survival and immunogenicity (Aim 1), and the role of HDAC11 in T-cell anti-melanoma immunity (Aim 2). In addition, the expertise provided by the Clinical Core will allow the successful completion of a Phase I clinical trial aimed to assess the safety and immunologic effects of HDAC inhibition in combination with an anti-CTLA4 antibody in patients with stage IV melanoma (Aim 3). The new knowledge to be generated by this team effort would lead to novel epigenetic-based immunotherapy that by overcoming the remarkable barrier of melanoma-induced T-cell tolerance would improve the efficacy of CTLA4 blockade.

Specific Aims:

In Specific Aim 1 we will characterize the functional effects of HDAC6 in melanoma cells to a) determine the effects of HDAC6 manipulation upon melanoma cell proliferation and/or survival, b) assess the effect of HDAC6 manipulation upon the immunogenicity of melanoma cells, c) mechanistically understand the role of HDAC6 and its molecular partner STAT3 in regulating cell proliferation/survival and immunogenicity of melanoma cells, and d) determine the in vivo anti-melanoma effect of HDAC6 specific inhibitor(s) alone or in combination with anti-CTLA4 antibodies. In Specific Aim 2 we will evaluate the role of HDAC11 in T-cell function as it relates to anti-melanoma immunity to a) determine the in vitro and in vivo function of melanoma-specific CD4 and CD8 T-cells lacking HDAC11, b) determine whether T-cells lacking HDAC11 are resistant to melanoma-induced T-cell tolerance in vivo, and c) determine the in vivo anti-melanoma effect of T-cells lacking HDAC11 either alone or in combination with anti-CTLA4 antibodies.

Finally, in Specific Aim 3, we will determine the safety and immunologic effects of epigenetic manipulation with HDAC inhibitors in combination with an anti-CTLA4 antibody in patients with stage IV melanoma performing a phase I study combining Panobinostat (LBH589) with Ipilimumab in patients with stage IV melanoma to assess the toxicities, immunologic and clinical effects of the combination.

CORE A: Tissue, Pathology and Bioinformatics

Core Co-Directors:
Jane Messina, M.D.
Steven Eschrich, Ph.D.

The Tissue, Pathology and Bioinformatics Core A of the Moffitt Skin SPORE will conduct the collection and storage of frozen tissue for lab-based translational studies (Projects 1, 2, and 3) as well as fixed tissue for molecular analyses and immunohistochemical staining/evaluation of molecular targets of interest (Projects 1, 2, and 3). The Core will storage and manage all data derived from these projects. The Core will also oversee the distribution of archived, clinically annotated biospecimens to support the Developmental Research and Career Development projects of the SPORE. More specifically, Core A of the Moffitt Skin SPORE will be a multifunctional core devoted to the (1) optimal procurement, handling, processing, and pathologic characterization of tissues from patients with melanoma and non-melanoma skin cancer, as well as the (2) accurate compilation, storage, and analysis of clinical, pathologic, and investigational data generated from the Skin SPORE projects. The ultimate goals of Core A are to utilize pathology and informatics expertise and resources to both support the translation of hypotheses generated by the basic science-driven projects into accurate, clinically relevant conclusions that can be generalized to the prevention and/or treatment of melanoma and non-melanoma skin cancer, and to carry out and/or support the translational assays that are included within the trials of this Skin SPORE. Core A will take advantage of and supplement institutional shared resources for tissue and blood procurement, banking, nucleic acid isolation, and histopathology services including immunohistochemistry and tissue microarray production, as well as analytic microscopy facilities for digital imaging and analysis. Pathologic review, grading, and classification of all patient tissue specimens will be conducted by Core A. Institutional information technology resources will provide researchers a centralized location to store data, access analytical tools, in a seamless application environment managed by the Core. These facilities will be leveraged by the co-leaders, who will oversee and manage acquisition, storage, and distribution of all tissues and data derived in the course of the projects.

Specific Aims:

There are four specific aims for The Tissue, Pathology and Bioinformatics Core A of the Moffitt Skin SPORE. Specific Aim 1 provides pathology services in support of the SPORE projects, including pathologic review of all patient material, tumor microstaging and classification, production of tissue microarrays, microdissection, immunohistochemistry, and DNA and RNA isolation. Specific Aim 2 establishes and tracks a repository of clinically- and pathologically-annotated tissue from patients with melanoma and other cutaneous malignancies in all stages of disease, to be used to support the projects encompassed by the Skin SPORE. Specific Aim 3 oversees and manages pathologic evaluation, quality assurance and quality control of tumor tissue generated for SPORE projects utilizing microscopy, digital imaging, and computer-assisted analysis. Finally, Specific Aim 4 is to be the SPORE data coordination center by providing services in data acquisition, transfer, integration, quality control, management and sharing for the Skin SPORE Projects and Cores and will a) coordinate data collection and transfer, perform quality control (QC) and facilitate data sharing, which involves developing software tools for managing and developing a standard data dictionary and quality control process, b) design, implement and maintain a centralized data management and computational system for the Skin SPORE projects that will facilitate acquisition, integration, quality control, analysis and sharing of the Skin SPORE projects’ data, including clinical data, specimen data, and assay results, and c) provide bioinformatics analysis support for genomics data generated by the Skin SPORE projects.

CORE B: Biostatistics

Core Co-Directors:
Michael Schell, Ph.D.
Ann Chen, Ph.D.

Biostatistics Core B will provide collaborative analytic support to all 3 Moffitt Skin SPORE projects, the other 2 Cores, and the Developmental Research and Career Development Programs. Of particular note, the core will provide statistical analytic activities of all three melanoma trials in Projects 1, 2, and 3. Each project has a faculty biostatistician, selected to maximize the link between their applied research focus and the project. There will be 3 clinical trials, including phase I trials in Projects 2 and 3 that will use a modified Ji design, chosen to provide a richer experience in establishing an MTD. This design, which received extensive discussion and included some modification by the Core B statisticians, will receive special ongoing attention as befits a novel analytical approach. Project 2 is very involved from an analytical perspective. Project 2 involves extensive proteomics analyses and uses a multi-level Bayesian model developed in part by the project 2 statistician. In conjunction with Cores A and C, this core will play a vital role in SPORE database development, especially with the clinical trials and case-cohort studies. Biostatistics Core B will be involved in the development of all proposed Developmental Research and Career Development Programs, and will provide analytic support to those that are funded. Working under the direction of faculty biostatisticians are two staff statisticians, who will handle a significant share of the direct statistical programming needed. Core members will be involved in all Projects at every stage of the research: Core members have worked closely with investigators to help them select efficient designs and adequate sample sizes to address study objectives. As studies proceed, Core B members will contribute to the refinement of standard operating procedures in response to realities of fieldwork, integrate data, monitor clinical trial databases, conduct data quality-control studies, manage compliance reporting, produce necessary reports, and conduct interim analyses. As the Projects yield results, the Core will conduct data analyses, prepare any necessary study reports, and assist investigators with the preparation of presentations and manuscripts. In addition, Core B will provide consultation on the design of subsequent research based on the experience of the current Projects. Core members will work closely with the Tissue, Pathology and Bioinformatics Core A and Administrative/Clinical Trials Core C for seamless integration of all aspects of this innovative Skin SPORE.

Specific Aims:

There are three specific aims of the Biostatistics Core B. Specific Aim 1 is to consult with Project investigators to select efficient study designs and appropriate sample sizes for the four research Projects, as well as DRP and CDP Projects in the Moffitt Skin SPORE. Specific Aim 2 is to support data management systems for clinical trials, including review and monitoring of clinical trials conducted within the Moffitt Skin SPORE project, thereby ensuring data quality, study integrity and patient safety. Finally, Specific Aim 3 is to prepare all summary statistics, provide up-to-date statistical analyses and assist in the preparation of abstracts and manuscripts that will disseminate the knowledge gained through the Moffitt Skin SPORE throughout the medical community.

CORE C: Administration and Clinical Trials

Core Co-Directors:
Jeffrey Weber, M.D., Ph.D.
James Mulé, Ph.D.

Administration and Clinical Trials Core C of the Moffitt Skin SPORE will be responsible for the successful carrying out of all SPORE activities related to financial oversight and coordination, publicity and record keeping in Projects 1, 2, and 3. The core will provide for integration of the regulatory and oversight activities of all three melanoma trials in Projects 1, 2, and 3 and ensure that the peer review, awarding and oversight processes of the Career Development and Developmental Research Programs are properly carried out. More specifically, the Administration and Clinical Trials Core C will serve to facilitate communication and manage interactions among SPORE investigators, and between SPORE members and other research and administrative units at Moffitt and with the NCI. Core C will provide organizational and financial oversight of all 3 projects, 3 cores and the Developmental Research and Career Development Programs, as well as provide for clinical trials coordination and data management support for all three melanoma trials integral to the Moffitt Skin SPORE. It includes support for a data manager and a clinical trials coordinator who will be critical to the performance of the trials of the SPORE. Administrative Core C also includes support for an administrator who will have overall day to day responsibility for the successful carrying out of all SPORE activities, including financial oversight and coordination, publicity and record keeping, and critically will coordinate the integration of the regulatory and oversight activities in the running of the Moffitt Skin SPORE. This will assure that all administrative requirements for successful completion of the overall SPORE goals and the requirements of the NCI and all other regulatory bodies including Moffitt, USF, the State of Florida and the FDA are met. Importantly, the Administrative Core C will coordinate the dissemination of information about the Developmental Research and Career Development Programs and oversee the reviews of those projects as well as the progress reports required. It will also provide for yearly meetings of the internal IAC and external EAB meetings. Core C will coordinate communication between the Moffitt Skin SPORE and other Skin Spores. It will provide the logistic capability necessary for development and support of all projects, cores, and programs, and it will integrate patient advocacy as a vital component of laboratory and clinical skin cancer research. Administrative Core C includes personnel who are vital to the successful function of the Moffitt Skin SPORE and are required across all projects.

Specific Aims:

The three specific aims of the Administration and Clinical Trials Core C of the Moffitt Skin SPORE are:

Specific Aim 1, General Administration and Communication: to provide organizational and financial oversight of all projects, cores, and programs; facilitate interdisciplinary research and collaboration among projects; to convene meetings with the external advisors, internal advisors, and foster frequent contacts and scientific exchanges with researchers within Moffitt interested in related research with particular focus on skin cancer, since open and free collaborations, communications, and exchange of resources are crucial to the success of the Moffitt Skin SPORE.

Specific Aim 2, Oversight and Regulation: Core C will allow investigators to devote maximal time to their research and minimize the time and effort devoted to organizational oversight and regulatory issues; to help prepare reports and continuation grant submissions to the NCI; maintain records on publications, presentations, and abstracts; and ensure adherence to regulatory requirements through interaction with the NCI, the Moffitt Cancer Center regulatory committees, the Institutional Review Board, Animal Use and Care Committee, and Biosafety Committees of the University of South Florida, the State of Florida and the FDA. Core C will also assist the Tissue, Pathology and Bioinformatics Core A with prioritization of tissue acquisition and will be responsible for oversight of clinical trials coordination, accrual to and data management of trials of the Moffitt Skin SPORE as well as trials prioritization.

Specific Aim 3, Patient Advocacy: to incorporate lay advocates into the advancement of skin cancer research, encourage and incorporate contributions of advocates in research design, implementation, and evaluation of the overall clinical trial process, and foster active participation of advocates in public education through institutional and community outreach.