1
|
Pillai M, Jolly MK. Systems-level network modeling deciphers the master regulators of phenotypic plasticity and heterogeneity in melanoma. iScience 2021; 24:103111. [PMID: 34622164 PMCID: PMC8479788 DOI: 10.1016/j.isci.2021.103111] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/03/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
Phenotypic (i.e. non-genetic) heterogeneity in melanoma drives dedifferentiation, recalcitrance to targeted therapy and immunotherapy, and consequent tumor relapse and metastasis. Various markers or regulators associated with distinct phenotypes in melanoma have been identified, but, how does a network of interactions among these regulators give rise to multiple "attractor" states and phenotypic switching remains elusive. Here, we inferred a network of transcription factors (TFs) that act as master regulators for gene signatures of diverse cell-states in melanoma. Dynamical simulations of this network predicted how this network can settle into different "attractors" (TF expression patterns), suggesting that TF network dynamics drives the emergence of phenotypic heterogeneity. These simulations can recapitulate major phenotypes observed in melanoma and explain de-differentiation trajectory observed upon BRAF inhibition. Our systems-level modeling framework offers a platform to understand trajectories of phenotypic transitions in the landscape of a regulatory TF network and identify novel therapeutic strategies targeting melanoma plasticity.
Collapse
Affiliation(s)
- Maalavika Pillai
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
- Undergraduate Programme, Indian Institute of Science, Bangalore, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
2
|
Bagaev A, Kotlov N, Nomie K, Svekolkin V, Gafurov A, Isaeva O, Osokin N, Kozlov I, Frenkel F, Gancharova O, Almog N, Tsiper M, Ataullakhanov R, Fowler N. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021; 39:845-865.e7. [PMID: 34019806 DOI: 10.1016/j.ccell.2021.04.014] [Citation(s) in RCA: 593] [Impact Index Per Article: 148.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/14/2020] [Accepted: 04/23/2021] [Indexed: 12/18/2022]
Abstract
The clinical use of molecular targeted therapy is rapidly evolving but has primarily focused on genomic alterations. Transcriptomic analysis offers an opportunity to dissect the complexity of tumors, including the tumor microenvironment (TME), a crucial mediator of cancer progression and therapeutic outcome. TME classification by transcriptomic analysis of >10,000 cancer patients identifies four distinct TME subtypes conserved across 20 different cancers. The TME subtypes correlate with patient response to immunotherapy in multiple cancers, with patients possessing immune-favorable TME subtypes benefiting the most from immunotherapy. Thus, the TME subtypes act as a generalized immunotherapy biomarker across many cancer types due to the inclusion of malignant and microenvironment components. A visual tool integrating transcriptomic and genomic data provides a global tumor portrait, describing the tumor framework, mutational load, immune composition, anti-tumor immunity, and immunosuppressive escape mechanisms. Integrative analyses plus visualization may aid in biomarker discovery and the personalization of therapeutic regimens.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nathan Fowler
- BostonGene, Waltham, MA 02453, USA; Department of Lymphoma and Myeloma, Unit 0429, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
3
|
Patel A, Carr MJ, Sun J, Zager JS. In-transit metastatic cutaneous melanoma: current management and future directions. Clin Exp Metastasis 2021; 39:201-211. [PMID: 33999365 DOI: 10.1007/s10585-021-10100-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
Management of in-transit melanoma encompasses a variety of possible treatment pathways and modalities. Depending on the location of disease, number of lesions, burden of disease and patient preference and characteristics, some treatments may be more beneficial than others. After full body radiographic staging is performed to rule out metastatic disease, curative therapy may be performed through surgical excision, intraarterial regional perfusion and infusion therapies, intralesional injections, systemic therapies or various combinations of any of these. While wide excision is limited in indication to superficial lesions that are few in number, the other listed therapies may be effective in treating unresectable disease. Where intraarterial perfusion based therapies have been shown to successfully treat extremity disease, injectable therapies can be used in lesions of the head and neck. Although systemic therapies for in-transit melanoma have limited specific data to support their primary use for in-transit disease, there are patients who may not be eligible for any of the other options, and current clinical trials are exploring the use of concurrent and sequential use of regional and systemic therapies with early results suggesting a synergistic benefit for oncologic response and outcomes.
Collapse
Affiliation(s)
- Ayushi Patel
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Michael J Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA
| | - James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA.,Department of Surgery, University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Zager
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA. .,Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
4
|
Peng D, Gleyzer R, Tai WH, Kumar P, Bian Q, Isaacs B, da Rocha EL, Cai S, DiNapoli K, Huang FW, Cahan P. Evaluating the transcriptional fidelity of cancer models. Genome Med 2021; 13:73. [PMID: 33926541 PMCID: PMC8086312 DOI: 10.1186/s13073-021-00888-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 04/15/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cancer researchers use cell lines, patient-derived xenografts, engineered mice, and tumoroids as models to investigate tumor biology and to identify therapies. The generalizability and power of a model derive from the fidelity with which it represents the tumor type under investigation; however, the extent to which this is true is often unclear. The preponderance of models and the ability to readily generate new ones has created a demand for tools that can measure the extent and ways in which cancer models resemble or diverge from native tumors. METHODS We developed a machine learning-based computational tool, CancerCellNet, that measures the similarity of cancer models to 22 naturally occurring tumor types and 36 subtypes, in a platform and species agnostic manner. We applied this tool to 657 cancer cell lines, 415 patient-derived xenografts, 26 distinct genetically engineered mouse models, and 131 tumoroids. We validated CancerCellNet by application to independent data, and we tested several predictions with immunofluorescence. RESULTS We have documented the cancer models with the greatest transcriptional fidelity to natural tumors, we have identified cancers underserved by adequate models, and we have found models with annotations that do not match their classification. By comparing models across modalities, we report that, on average, genetically engineered mice and tumoroids have higher transcriptional fidelity than patient-derived xenografts and cell lines in four out of five tumor types. However, several patient-derived xenografts and tumoroids have classification scores that are on par with native tumors, highlighting both their potential as faithful model classes and their heterogeneity. CONCLUSIONS CancerCellNet enables the rapid assessment of transcriptional fidelity of tumor models. We have made CancerCellNet available as a freely downloadable R package ( https://github.com/pcahan1/cancerCellNet ) and as a web application ( http://www.cahanlab.org/resources/cancerCellNet_web ) that can be applied to new cancer models that allows for direct comparison to the cancer models evaluated here.
Collapse
Affiliation(s)
- Da Peng
- grid.21107.350000 0001 2171 9311Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Rachel Gleyzer
- grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Wen-Hsin Tai
- grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Pavithra Kumar
- grid.21107.350000 0001 2171 9311Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ,grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Qin Bian
- grid.21107.350000 0001 2171 9311Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ,grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Bradley Isaacs
- grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Edroaldo Lummertz da Rocha
- grid.411237.20000 0001 2188 7235Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, SC Brazil
| | - Stephanie Cai
- grid.21107.350000 0001 2171 9311Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Kathleen DiNapoli
- grid.21107.350000 0001 2171 9311Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ,grid.21107.350000 0001 2171 9311Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD 21218 USA
| | - Franklin W. Huang
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine; Helen Diller Family Cancer Center; Bakar Computational Health Sciences Institute; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA USA
| | - Patrick Cahan
- grid.21107.350000 0001 2171 9311Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ,grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ,grid.21107.350000 0001 2171 9311Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
5
|
Perone JA, Farrow N, Tyler DS, Beasley GM. Contemporary Approaches to In-Transit Melanoma. J Oncol Pract 2019; 14:292-300. [PMID: 29746804 DOI: 10.1200/jop.18.00063] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In-transit melanoma represents a distinct disease pattern of heterogeneous superficial tumors. Many treatments have been developed specifically for this type of disease, including regional chemotherapy and a variety of directly injectable agents. Novel strategies include the intralesional delivery of oncolytic viruses and immunocytokines. The combination of intralesional or regional chemotherapy with systemic immune checkpoint inhibitors also is a promising approach. In the current review, we examine the general management of the workup of patients with in-transit disease, the range of available therapies, and recommendations for specific therapies for an individual patient.
Collapse
Affiliation(s)
- Jennifer A Perone
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Nellie Farrow
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Douglas S Tyler
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Georgia M Beasley
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| |
Collapse
|
6
|
Jeanne A, Boulagnon-Rombi C, Devy J, Théret L, Fichel C, Bouland N, Diebold MD, Martiny L, Schneider C, Dedieu S. Matricellular TSP-1 as a target of interest for impeding melanoma spreading: towards a therapeutic use for TAX2 peptide. Clin Exp Metastasis 2016; 33:637-49. [PMID: 27349907 DOI: 10.1007/s10585-016-9803-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/27/2016] [Indexed: 01/07/2023]
Abstract
Thrombospondin-1 (TSP-1) is a matricellular glycoprotein known for being highly expressed within a tumor microenvironment, where it promotes an aggressive phenotype particularly by interacting with the CD47 cell-surface receptor. While it originates from the stromal compartment in many malignancies, melanoma is an exception as invasive and metastatic melanoma cells overexpress TSP-1. We recently demonstrated that a new molecular agent that selectively prevents TSP-1 binding to CD47, called TAX2, exhibits anti-cancer properties when administered systemically by decreasing viable tumor tissue within subcutaneous B16 melanoma allografts. At the same time, emerging evidence was published suggesting a contribution of TSP-1 in melanoma metastatic dissemination and resistance to treatment. Through a comprehensive systems biology approach based on multiple genomics and proteomics databases analyses, we first identified a TSP-1-centered interaction network that is overexpressed in metastatic melanoma. Then, we investigated the effects of disrupting TSP-1:CD47 interaction in A375 human malignant melanoma xenografts. In this model, TAX2 systemic administrations induce tumor necrosis by decreasing intra-tumoral blood flow, while concomitantly making tumors less infiltrative. Besides, TAX2 treatment also drastically inhibits B16F10 murine melanoma cells metastatic dissemination and growth in a syngeneic experimental model of lung metastasis, as demonstrated by histopathological analyses as well as longitudinal and quantitative µCT follow-up of metastatic progression. Altogether, the results obtained by combining bioinformatics and preclinical studies strongly suggest that targeting TSP-1/CD47 axis may represent a valuable therapeutic alternative for hampering melanoma spreading.
Collapse
Affiliation(s)
- Albin Jeanne
- Laboratoire SiRMa, Campus Moulin de La Housse, Université de Reims Champagne-Ardenne (URCA), UFR Sciences Exactes Et Naturelles, BP 1039, 51687, Reims Cedex 2, France
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
- SATT Nord, Lille, France
| | - Camille Boulagnon-Rombi
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
- CHU de Reims, Laboratoire Central D'Anatomie Et de Cytologie Pathologiques, Reims, France
| | - Jérôme Devy
- Laboratoire SiRMa, Campus Moulin de La Housse, Université de Reims Champagne-Ardenne (URCA), UFR Sciences Exactes Et Naturelles, BP 1039, 51687, Reims Cedex 2, France
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
| | - Louis Théret
- Laboratoire SiRMa, Campus Moulin de La Housse, Université de Reims Champagne-Ardenne (URCA), UFR Sciences Exactes Et Naturelles, BP 1039, 51687, Reims Cedex 2, France
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
| | - Caroline Fichel
- Université de Reims Champagne-Ardenne, Laboratoire D'Anatomie Pathologique, UFR Médecine, Reims, France
| | - Nicole Bouland
- Université de Reims Champagne-Ardenne, Laboratoire D'Anatomie Pathologique, UFR Médecine, Reims, France
| | - Marie-Danièle Diebold
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
- CHU de Reims, Laboratoire Central D'Anatomie Et de Cytologie Pathologiques, Reims, France
| | - Laurent Martiny
- Laboratoire SiRMa, Campus Moulin de La Housse, Université de Reims Champagne-Ardenne (URCA), UFR Sciences Exactes Et Naturelles, BP 1039, 51687, Reims Cedex 2, France
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
| | - Christophe Schneider
- Laboratoire SiRMa, Campus Moulin de La Housse, Université de Reims Champagne-Ardenne (URCA), UFR Sciences Exactes Et Naturelles, BP 1039, 51687, Reims Cedex 2, France
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France
| | - Stéphane Dedieu
- Laboratoire SiRMa, Campus Moulin de La Housse, Université de Reims Champagne-Ardenne (URCA), UFR Sciences Exactes Et Naturelles, BP 1039, 51687, Reims Cedex 2, France.
- CNRS UMR 7369, Unité Matrice Extracellulaire Et Dynamique Cellulaire, MEDyC, Reims, France.
| |
Collapse
|
7
|
Lawson MA, McDonald MM, Kovacic N, Hua Khoo W, Terry RL, Down J, Kaplan W, Paton-Hough J, Fellows C, Pettitt JA, Neil Dear T, Van Valckenborgh E, Baldock PA, Rogers MJ, Eaton CL, Vanderkerken K, Pettit AR, Quinn JMW, Zannettino ACW, Phan TG, Croucher PI. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat Commun 2015; 6:8983. [PMID: 26632274 PMCID: PMC4686867 DOI: 10.1038/ncomms9983] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/23/2015] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma is largely incurable, despite development of therapies that target myeloma cell-intrinsic pathways. Disease relapse is thought to originate from dormant myeloma cells, localized in specialized niches, which resist therapy and repopulate the tumour. However, little is known about the niche, and how it exerts cell-extrinsic control over myeloma cell dormancy and reactivation. In this study, we track individual myeloma cells by intravital imaging as they colonize the endosteal niche, enter a dormant state and subsequently become activated to form colonies. We demonstrate that dormancy is a reversible state that is switched ‘on' by engagement with bone-lining cells or osteoblasts, and switched ‘off' by osteoclasts remodelling the endosteal niche. Dormant myeloma cells are resistant to chemotherapy that targets dividing cells. The demonstration that the endosteal niche is pivotal in controlling myeloma cell dormancy highlights the potential for targeting cell-extrinsic mechanisms to overcome cell-intrinsic drug resistance and prevent disease relapse. Therapy resistant dormant myeloma cells contribute to disease relapse. Here, the authors use intravital microscopy to track the location of these cells and demonstrate that they hone to the endosteal niche within the bone.
Collapse
Affiliation(s)
- Michelle A Lawson
- Department of Oncology, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK
| | - Michelle M McDonald
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Natasa Kovacic
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia
| | - Weng Hua Khoo
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,School of Biotechnology and Biomolecular Sciences, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Rachael L Terry
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Jenny Down
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia
| | - Warren Kaplan
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Julia Paton-Hough
- Department of Oncology, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK
| | - Clair Fellows
- Department of Oncology, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK
| | - Jessica A Pettitt
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia
| | - T Neil Dear
- South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Paul A Baldock
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Michael J Rogers
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Colby L Eaton
- Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK.,Department of Human Metabolism and Clinical Biochemistry, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield, South Yorkshire S10 2RX, UK
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Allison R Pettit
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Julian M W Quinn
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia
| | - Andrew C W Zannettino
- South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,School of Medical Sciences, University of Adelaide, Frome Road, Adelaide, South Australia 5000, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, New South Wales 2010, Australia
| |
Collapse
|
8
|
Svetlichnyy D, Imrichova H, Fiers M, Kalender Atak Z, Aerts S. Identification of High-Impact cis-Regulatory Mutations Using Transcription Factor Specific Random Forest Models. PLoS Comput Biol 2015; 11:e1004590. [PMID: 26562774 PMCID: PMC4642938 DOI: 10.1371/journal.pcbi.1004590] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 10/10/2015] [Indexed: 02/02/2023] Open
Abstract
Cancer genomes contain vast amounts of somatic mutations, many of which are passenger mutations not involved in oncogenesis. Whereas driver mutations in protein-coding genes can be distinguished from passenger mutations based on their recurrence, non-coding mutations are usually not recurrent at the same position. Therefore, it is still unclear how to identify cis-regulatory driver mutations, particularly when chromatin data from the same patient is not available, thus relying only on sequence and expression information. Here we use machine-learning methods to predict functional regulatory regions using sequence information alone, and compare the predicted activity of the mutated region with the reference sequence. This way we define the Predicted Regulatory Impact of a Mutation in an Enhancer (PRIME). We find that the recently identified driver mutation in the TAL1 enhancer has a high PRIME score, representing a “gain-of-target” for MYB, whereas the highly recurrent TERT promoter mutation has a surprisingly low PRIME score. We trained Random Forest models for 45 cancer-related transcription factors, and used these to score variations in the HeLa genome and somatic mutations across more than five hundred cancer genomes. Each model predicts only a small fraction of non-coding mutations with a potential impact on the function of the encompassing regulatory region. Nevertheless, as these few candidate driver mutations are often linked to gains in chromatin activity and gene expression, they may contribute to the oncogenic program by altering the expression levels of specific oncogenes and tumor suppressor genes. Precise regulation of gene expression is controlled by cis-regulatory modules (CRM) containing binding sites for transcription factors (TF). The genome-wide location of all TF binding sites can often be obtained by ChIP-seq (chromatin immunoprecipitation followed by deep sequencing), yet in most cases only a minority of the binding peaks actually represent functional CRMs that control the transcription initiation of a bona fide TF target gene. Here, we investigated for 45 cancer-related TFs how machine-learning approaches can be used to predict functional TF target CRMs. After careful evaluation of their performance, we used these TF-target classifiers to predict which cis-regulatory mutations may have a significant impact on gene regulation by evaluating whether the mutation causes a significant gain or loss in the probability that the CRM is a functional TF target. We found that Random Forest classifiers can achieve more than 100-fold higher specificity for mutation prediction compared to the simple approaches based on scanning with position weight matrices. By scanning somatic mutations in breast cancer genomes and in the HeLa genome, we finally show that our TF-target classifiers can identify high impact non-coding mutations that are associated with concordant TF binding, gene expression changes and chromatin activity. In conclusion, TF-specific Random Forest classifiers can be used to prioritize cis-regulatory mutations in cancer genomes with high accuracy.
Collapse
Affiliation(s)
- Dmitry Svetlichnyy
- Laboratory of Computational Biology, KU Leuven Center for Human Genetics, Leuven, Belgium
| | - Hana Imrichova
- Laboratory of Computational Biology, KU Leuven Center for Human Genetics, Leuven, Belgium
| | - Mark Fiers
- VIB Center for the Biology of Disease, Leuven, Belgium
| | - Zeynep Kalender Atak
- Laboratory of Computational Biology, KU Leuven Center for Human Genetics, Leuven, Belgium
| | - Stein Aerts
- Laboratory of Computational Biology, KU Leuven Center for Human Genetics, Leuven, Belgium
- * E-mail:
| |
Collapse
|
9
|
Hassan S, Petrella TM, Zhang T, Kamel-Reid S, Nordio F, Baccarelli A, Sade S, Naert K, Habeeb AA, Ghazarian D, Wright FC. Pathologic complete response to intralesional interleukin-2 therapy associated with improved survival in melanoma patients with in-transit disease. Ann Surg Oncol 2014; 22:1950-8. [PMID: 25366584 DOI: 10.1245/s10434-014-4199-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Indexed: 12/14/2022]
Abstract
PURPOSE Melanoma patients with in-transit disease have a high mortality rate despite various treatment strategies. The aim of this study was to validate the role of intralesional interleukin (IL)-2, to understand its mechanism of action, and to better understand factors that may influence its response. METHODS We retrospectively collected the clinicopathological data of 31 consecutive patients who presented to a tertiary care cancer center for treatment of in-transit melanoma with intralesional IL-2. Kaplan-Meier survival curves and multivariable Cox regression analysis were performed. Immunohistochemistry (IHC) was used to better understand the immune response to localized IL-2 therapy. Targeted next-generation sequencing was performed to genomically characterize the tumors. RESULTS Ten patients (10/31, 32 %) achieved a pathologic complete response (pCR), 17/21 (55 %) had a partial response, and 4/21 (19 %) had progressive disease on treatment. pCR to IL-2 therapy was associated with overall survival (log-rank p = 0.004) and improved progression-free survival (PFS) [adjusted hazard ratio (HR) 0.11; 95 % CI 0.02-0.47; p = 0.003). A higher CD8+ T cell infiltrate was identified in in-transit lesions with a pCR compared with the other lesions (mean IHC score 3.78 vs. 2.61; p = 0.01). Patients with an elevated CD8+ infiltrate demonstrated an improved PFS (unadjusted HR 0.08; 95 % CI 0.01-0.52; p = 0.008). CONCLUSIONS Thirty-two percent of patients achieved pCR with intralesional IL-2 therapy and had a significantly improved PFS compared with the rest of the cohort, which may be explained by a systemic CD8+ T-cell response.
Collapse
Affiliation(s)
- Saima Hassan
- Division of Surgical Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Tokuhisa Y, Lidsky ME, Toshimitsu H, Turley RS, Beasley GM, Ueno T, Sharma K, Augustine CK, Tyler DS. SRC family kinase inhibition as a novel strategy to augment melphalan-based regional chemotherapy of advanced extremity melanoma. Ann Surg Oncol 2013; 21:1024-30. [PMID: 24281418 DOI: 10.1245/s10434-013-3387-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Indexed: 11/18/2022]
Abstract
BACKGROUND Src kinase inhibition has been shown to augment the efficacy of chemotherapy. Dasatinib, a dual Src/Abl kinase inhibitor approved for the treatment of CML, is under investigation as monotherapy for tumors with abnormal Src signaling, such as melanoma. The goal of this study was to determine if Src kinase inhibition using dasatinib could enhance the efficacy of regionally administered melphalan in advanced extremity melanoma. METHODS The mutational status of c-kit and patterns of gene expression predictive of dysregulated Src kinase signaling were evaluated in a panel of 26 human melanoma cell lines. The effectiveness of dasatinib was measured by quantifying protein expression and activation of Src kinase, focal adhesion kinase, and Crk-associated substrate (p130(CAS)), in conjunction with in vitro cell viability assays using seven melanoma cell lines. Utilizing a rat model of regional chemotherapy, we evaluated the effectiveness of systemic dasatinib in conjunction with regional melphalan against the human melanoma cell line, DM443, grown as a xenograft. RESULTS Only the WM3211 cell line harbored a c-kit mutation. Significant correlation was observed between Src-predicted dysregulation by gene expression and sensitivity to dasatinib in vitro. Tumor doubling time for DM443 xenografts treated with systemic dasatinib in combination with regional melphalan (44.8 days) was significantly longer (p = 0.007) than either dasatinib (21.3 days) or melphalan alone (24.7 days). CONCLUSIONS Systemic dasatinib prior to melphalan-based regional chemotherapy markedly improves the efficacy of this alkylating agent in this melanoma xenograft model. Validation of this concept should be considered in the context of a regional therapy clinical trial.
Collapse
|
11
|
Lidsky ME, Speicher PJ, Jiang B, Tsutsui M, Tyler DS. Isolated limb infusion as a model to test new agents to treat metastatic melanoma. J Surg Oncol 2013; 109:357-65. [PMID: 24522940 DOI: 10.1002/jso.23502] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/24/2013] [Indexed: 02/06/2023]
Abstract
The limb model of in-transit disease can expand our understanding of treating melanoma because of the ease of obtaining tissue biopsies for correlative studies and the availability of preclinical animal models that allow validation of novel therapeutic strategies. This review will focus on regional therapy for in-transit melanoma as a platform to investigate novel therapeutic approaches to improve regional disease control, and help us develop insights to more rationally design systemic therapy trials.
Collapse
Affiliation(s)
- Michael E Lidsky
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | |
Collapse
|
12
|
Hanks BA, Holtzhausen A, Evans KS, Jamieson R, Gimpel P, Campbell OM, Hector-Greene M, Sun L, Tewari A, George A, Starr M, Nixon A, Augustine C, Beasley G, Tyler DS, Osada T, Morse MA, Ling L, Lyerly HK, Blobe GC. Type III TGF-β receptor downregulation generates an immunotolerant tumor microenvironment. J Clin Invest 2013; 123:3925-40. [PMID: 23925295 PMCID: PMC3754240 DOI: 10.1172/jci65745] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 06/13/2013] [Indexed: 01/02/2023] Open
Abstract
Cancers subvert the host immune system to facilitate disease progression. These evolved immunosuppressive mechanisms are also implicated in circumventing immunotherapeutic strategies. Emerging data indicate that local tumor-associated DC populations exhibit tolerogenic features by promoting Treg development; however, the mechanisms by which tumors manipulate DC and Treg function in the tumor microenvironment remain unclear. Type III TGF-β receptor (TGFBR3) and its shed extracellular domain (sTGFBR3) regulate TGF-β signaling and maintain epithelial homeostasis, with loss of TGFBR3 expression promoting progression early in breast cancer development. Using murine models of breast cancer and melanoma, we elucidated a tumor immunoevasion mechanism whereby loss of tumor-expressed TGFBR3/sTGFBR3 enhanced TGF-β signaling within locoregional DC populations and upregulated both the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO) in plasmacytoid DCs and the CCL22 chemokine in myeloid DCs. Alterations in these DC populations mediated Treg infiltration and the suppression of antitumor immunity. Our findings provide mechanistic support for using TGF-β inhibitors to enhance the efficacy of tumor immunotherapy, indicate that sTGFBR3 levels could serve as a predictive immunotherapy biomarker, and expand the mechanisms by which TGFBR3 suppresses cancer progression to include effects on the tumor immune microenvironment.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Chemokine CCL22/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Down-Regulation
- Female
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Proteoglycans/genetics
- Proteoglycans/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/metabolism
- Tumor Escape
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Brent A. Hanks
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Alisha Holtzhausen
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Katherine S. Evans
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Rebekah Jamieson
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Petra Gimpel
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Olivia M. Campbell
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Melissa Hector-Greene
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Lihong Sun
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Alok Tewari
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Amanda George
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Mark Starr
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Andrew Nixon
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Christi Augustine
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia Beasley
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Douglas S. Tyler
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Takayu Osada
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Michael A. Morse
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Leona Ling
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - H. Kim Lyerly
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Gerard C. Blobe
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
13
|
Shetty G, Beasley GM, Sparks S, Barfield M, Masoud M, Mosca PJ, Pruitt SK, Salama AKS, Chan C, Tyler DS, Weinhold KJ. Plasma cytokine analysis in patients with advanced extremity melanoma undergoing isolated limb infusion. Ann Surg Oncol 2013; 20:1128-35. [PMID: 23456379 DOI: 10.1245/s10434-012-2785-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND Preprocedure clinical and pathologic factors have failed to consistently differentiate complete response (CR) from progressive disease (PD) in patients after isolated limb infusion (ILI) with melphalan for unresectable in-transit extremity melanoma. METHODS Multiplex immunobead assay technology (Milliplex MAP Human Cytokine/Chemokine Magnetic Bead Panel, Millipore Corp., Billerica, MA; and Magpix analytical test instrument, Luminex Corp., Austin, TX) was performed on pre-ILI plasma to determine concentrations of selected cytokines (MIP-1α, IL-1Rα, IP-10, IL-1β, IL-1α, MCP-1, IL-6, IL-17, EGF, IL-12p40, VEGF, GM-CSF, and MIP-1β) on a subset of patients (n = 180) who experienced CR (n = 23) or PD (n = 24) after ILI. Plasma from normal donors (n = 12) was also evaluated. RESULTS Of 180 ILIs performed, 28 % (95 % confidence interval 22-35, n = 50) experienced a CR, 14 % (n = 25) experienced a partial response, 11 % (n = 21) had stable disease, 34 % (n = 61) had PD, and 13 % (n = 23) were not evaluable for response. Tumor characteristics and pharmacokinetics appeared similar between CR (n = 23) and PD (n = 24) patients who underwent cytokine analysis. Although there were no differences in cytokine levels between CR and PD patients, there were differences between the melanoma patients and controls. MIP-1α, IL-1Rα, IL-1β, IL-1α, IL-17, EGF, IL-12p40, VEGF, GM-CSF, and MIP-1β were significantly higher in normal controls compared to melanoma patients, while IP-10 was lower (p < 0.001) in controls compared to melanoma patients. CONCLUSIONS Patients with unresectable in-transit melanoma appear to have markedly decreased levels of immune activating cytokines compared to normal healthy controls. This further supports a potential role for immune-targeted therapies and immune monitoring in patients with regionally advanced melanoma.
Collapse
Affiliation(s)
- Gina Shetty
- Department of Immunology, Duke University, Durham, NC, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jewell R, Mitra A, Conway C, Iremonger J, Walker C, de Kort F, Cook M, Boon A, Speirs V, Newton-Bishop J. Identification of differentially expressed genes in matched formalin-fixed paraffin-embedded primary and metastatic melanoma tumor pairs. Pigment Cell Melanoma Res 2012; 25:284-6. [DOI: 10.1111/j.1755-148x.2011.00965.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Freedman JA, Tyler DS, Nevins JR, Augustine CK. Use of gene expression and pathway signatures to characterize the complexity of human melanoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2513-22. [PMID: 21641377 DOI: 10.1016/j.ajpath.2011.02.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/03/2011] [Accepted: 02/14/2011] [Indexed: 11/29/2022]
Abstract
A defining characteristic of most human cancers is heterogeneity, resulting from the somatic acquisition of a complex array of genetic and genomic alterations. Dissecting this heterogeneity is critical to developing an understanding of the underlying mechanisms of disease and to paving the way toward personalized treatments of the disease. We used gene expression data sets from the analysis of primary and metastatic melanomas to develop a molecular description of the heterogeneity that characterizes this disease. Unsupervised hierarchical clustering, gene set enrichment analyses, and pathway activity analyses were used to describe the genetic heterogeneity of melanomas. Patterns of gene expression that revealed two distinct classes of primary melanoma, two distinct classes of in-transit melanoma, and at least three subgroups of metastatic melanoma were identified. Expression signatures developed to predict the status of oncogenic signaling pathways were used to explore the biological basis underlying these differential patterns of expression. This analysis of activities revealed unique pathways that distinguished the primary and metastatic subgroups of melanoma. Distinct patterns of gene expression across primary, in-transit, and metastatic melanomas underline the genetic heterogeneity of this disease. This heterogeneity can be described in terms of deregulation of signaling pathways, thus increasing the knowledge of the biological features underlying individual melanomas and potentially directing therapeutic opportunities to individual patients with melanoma.
Collapse
Affiliation(s)
- Jennifer A Freedman
- Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | |
Collapse
|
16
|
Freedman JA, Augustine CK, Selim AM, Holshausen KC, Wei Z, Tsamis KA, Hsu DS, Dressman HK, Barry WT, Tyler DS, Nevins JR. A methodology for utilization of predictive genomic signatures in FFPE samples. BMC Med Genomics 2011; 4:58. [PMID: 21745407 PMCID: PMC3146808 DOI: 10.1186/1755-8794-4-58] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 07/11/2011] [Indexed: 01/05/2023] Open
Abstract
Background Gene expression signatures developed to measure the activity of oncogenic signaling pathways have been used to dissect the heterogeneity of tumor samples and to predict sensitivity to various cancer drugs that target components of the relevant pathways, thus potentially identifying therapeutic options for subgroups of patients. To facilitate broad use, including in a clinical setting, the ability to generate data from formalin-fixed, paraffin-embedded (FFPE) tissues is essential. Methods Patterns of pathway activity in matched fresh-frozen and FFPE xenograft tumor samples were generated using the MessageAmp Premier methodology in combination with assays using Affymetrix arrays. Results generated were compared with those obtained from fresh-frozen samples using a standard Affymetrix assay. In addition, gene expression data from patient matched fresh-frozen and FFPE melanomas were also utilized to evaluate the consistency of predictions of oncogenic signaling pathway status. Results Significant correlation was observed between pathway activity predictions from paired fresh-frozen and FFPE xenograft tumor samples. In addition, significant concordance of pathway activity predictions was also observed between patient matched fresh-frozen and FFPE melanomas. Conclusions Reliable and consistent predictions of oncogenic pathway activities can be obtained from FFPE tumor tissue samples. The ability to reliably utilize FFPE patient tumor tissue samples for genomic analyses will lead to a better understanding of the biology of disease progression and, in the clinical setting, will provide tools to guide the choice of therapeutics to those most likely to be effective in treating a patient's disease.
Collapse
Affiliation(s)
- Jennifer A Freedman
- Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, NC 27708, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Nardin A, Wong WC, Tow C, Molina TJ, Tissier F, Audebourg A, Garcette M, Caignard A, Avril MF, Abastado JP, Prévost-Blondel A. Dacarbazine promotes stromal remodeling and lymphocyte infiltration in cutaneous melanoma lesions. J Invest Dermatol 2011; 131:1896-905. [PMID: 21654834 DOI: 10.1038/jid.2011.128] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dacarbazine (DTIC) is the standard first-line drug for advanced stage melanoma, but it induces objective clinical responses in only 15% of patients. This study was designed to identify molecular changes specifically induced by treatment in chemo-sensitive lesions. Using global transcriptome analysis and immunohistochemistry, we analyzed cutaneous metastases resected from patients with melanoma before and after DTIC treatment. The treatment induced similar functional changes in different lesions from the same patient. Stromal and immune response-related genes were the most frequently upregulated, particularly in lesions that responded to treatment by stabilizing or regressing. T-cell infiltration and enhanced major histocompatibility complex class II expression were observed in a subset of patients. Stable, chemo-sensitive lesions exhibited activation of genetic programs related to extracellular matrix remodeling, including increased expression of secreted protein acidic and rich in cysteine (SPARC) by tumor cells. These events were associated with local response to treatment and with superior survival in our group of patients. In contrast, SPARC expression was downregulated in lesions resistant to DTIC. Thus, chemotherapy drugs originally selected for their direct cytotoxicity to tumor cells may also influence disease progression by inducing changes in the tumor microenvironment.
Collapse
|
18
|
Abstract
For in-transit melanoma confined to the extremities, regional chemotherapy in the form of hyperthermic isolated limb perfusion and isolated limb infusion are effective treatment modalities carrying superior response rates to current standard systemic therapy. Despite high response rates, most patients will eventually recur, supporting the role for novel research aimed at improving durable responses and minimizing toxicity. Although the standard cytotoxic agent for regional chemotherapy is melphalan, alternative agents such as temozolomide are currently being tested, with promising preliminary results. Current strategies for improving chemosensitivity to regional chemotherapy are aimed at overcoming classic resistance mechanisms such as drug metabolism and DNA repair, increasing drug delivery, inhibiting tumor-specific angiogenesis, and decreasing the apoptotic threshold of melanoma cells. Concurrent with development and testing of these agents, genomic profiling and biomolecular analysis of acquired tumor tissue may define patterns of tumor resistance and sensitivity from which personalized treatment may be tailored to optimize efficacy. In this article rational strategies for treatment of in-transit melanoma are outlined, with special emphasis on current translational and clinical research efforts.
Collapse
|
19
|
Beasley GM, Riboh JC, Augustine CK, Zager JS, Hochwald SN, Grobmyer SR, Peterson B, Royal R, Ross MI, Tyler DS. Prospective multicenter phase II trial of systemic ADH-1 in combination with melphalan via isolated limb infusion in patients with advanced extremity melanoma. J Clin Oncol 2011; 29:1210-5. [PMID: 21343562 DOI: 10.1200/jco.2010.32.1224] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Isolated limb infusion (ILI) with melphalan (M-ILI) dosing corrected for ideal body weight (IBW) is a well-tolerated treatment for patients with in-transit melanoma with a 29% complete response rate. ADH-1 is a cyclic pentapeptide that disrupts N-cadherin adhesion complexes. In a preclinical animal model, systemic ADH-1 given with regional melphalan demonstrated synergistic antitumor activity, and in a phase I trial with M-ILI it had minimal toxicity. PATIENTS AND METHODS Patients with American Joint Committee on Cancer (AJCC) stage IIIB or IIIC extremity melanoma were treated with 4,000 mg of ADH-1, administered systemically on days 1 and 8, and with M-ILI corrected for IBW on day 1. Drug pharmacokinetics and N-cadherin immunohistochemical staining were performed on pretreatment tumor. The primary end point was response at 12 weeks determined by Response Evaluation Criteria in Solid Tumors (RECIST) criteria. RESULTS In all, 45 patients were enrolled over 15 months at four institutions. In-field responses included 17 patients with complete responses (CRs; 38%), 10 with partial responses (22%), six with stable disease (13%), eight with progressive disease (18%), and four (9%) who were not evaluable. Median duration of in-field response among the 17 CRs was 5 months, and median time to in-field progression among 41 evaluable patients was 4.6 months (95% CI, 4.0 to 7.1 months). N-cadherin was detected in 20 (69%) of 29 tumor samples. Grade 4 toxicities included creatinine phosphokinase increase (four patients), arterial injury (one), neutropenia (one), and pneumonitis (one). CONCLUSION To the best of our knowledge, this phase II trial is the first prospective multicenter ILI trial and the first to incorporate a targeted agent in an attempt to augment antitumor responses to regional chemotherapy. Although targeting N-cadherin may improve melanoma sensitivity to chemotherapy, no difference in response to treatment was seen in this study.
Collapse
|