101
|
Wu S, Wang Y, Sun L, Zhang Z, Jiang Z, Qin Z, Han H, Liu Z, Li X, Tang A, Gui Y, Cai Z, Zhou F. Decreased expression of dual-specificity phosphatase 9 is associated with poor prognosis in clear cell renal cell carcinoma. BMC Cancer 2011; 11:413. [PMID: 21943117 PMCID: PMC3198720 DOI: 10.1186/1471-2407-11-413] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2011] [Accepted: 09/26/2011] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The molecular mechanisms involved in the development and progression of clear cell renal cell carcinomas (ccRCCs) are poorly understood. The objective of this study was to analyze the expression of dual-specificity phosphatase 9 (DUSP-9) and determine its clinical significance in human ccRCCs. METHODS The expression of DUSP-9 mRNA was determined in 46 paired samples of ccRCCs and adjacent normal tissues by using real-time qPCR. The expression of the DUSP-9 was determined in 211 samples of ccRCCs and 107 paired samples of adjacent normal tissues by immunohistochemical analysis. Statistical analysis was performed to define the relationship between the expression of DUSP-9 and the clinical features of ccRCC. RESULTS The mRNA level of DUSP-9, which was determined by real-time RT-PCR, was found to be significantly lower in tumorous tissues than in the adjacent non-tumorous tissues (p < 0.001). An immunohistochemical analysis of 107 paired tissue specimens showed that the DUSP-9 expression was lower in tumorous tissues than in the adjacent non-tumorous tissues (p < 0.001). Moreover, there was a significant correlation between the DUSP-9 expression in ccRCCs and gender (p = 0.031), tumor size (p = 0.001), pathologic stage (p = 0.001), Fuhrman grade (p = 0.002), T stage (p = 0.001), N classification (p = 0.012), metastasis (p = 0.005), and recurrence (p < 0.001). Patients with lower DUSP-9 expression had shorter overall survival time than those with higher DUSP-9 expression (p < 0.001). Multivariate analysis indicated that low expression of the DUSP-9 was an independent predictor for poor survival of ccRCC patients. CONCLUSION To our knowledge, this is the first study that determines the relationship between DUSP-9 expression and prognosis in ccRCC. We found that decreased expression of DUSP-9 is associated with poor prognosis in ccRCC. DUSP-9 may represent a novel and useful prognostic marker for ccRCC.
Collapse
Affiliation(s)
- Song Wu
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Tarhini AA, Frankel P, Margolin KA, Christensen S, Ruel C, Shipe-Spotloe J, Gandara DR, Chen A, Kirkwood JM. Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin Cancer Res 2011; 17:6574-81. [PMID: 21880788 DOI: 10.1158/1078-0432.ccr-11-1463] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aflibercept is a soluble decoy VEGF receptor and angiogenesis inhibitor with potent preclinical antitumor activity in melanoma. We conducted a multicenter phase II study in patients with inoperable stage III or IV melanoma and no prior chemotherapy. EXPERIMENTAL DESIGN A two-stage design was adopted to evaluate 4-month progression-free survival rate (PFSR) and response rate. Aflibercept was given at 4 mg/kg intravenously every 2 weeks. Response was assessed every 8 weeks. First-stage accrual of 21 patients was specified and with an adequate 4-month PFSR accrual continued to a total of 41. RESULTS Forty-one patients of ages 23 to 84 (median = 57) were enrolled. Thirty-nine had American Joint Committee on Cancer stage IV (5 M1a, 7 M1b, and 27 M1c) and 2 had inoperable stage IIIC (N3). Eastern Cooperative Oncology Group (ECOG) performance status was 0 (27 patients) or 1 (14 patients). Ten patients had primary uveal melanoma, 28 cutaneous, and 3 had unknown primaries. A median of 7 cycles were initiated (range: 1-56). Grade 3 and 4 toxicities included hypertension in 9 patients (22%) and proteinuria in 6 (15%). Among 40 patients evaluable for efficacy (those who initiated aflibercept), 3 (7.5%) had a confirmed partial response and 20 had progression-free survival of 4 months or above. The predicted 1-year survival rate derived from the Korn meta-analysis model is 36% (N = 39), whereas we observed a corresponding 56.4% survival rate at 1 year (95% CI, 43-74, P < 0.005). Median overall survival in this trial is 16.3 months (95% CI, 9.2 to not reached). We observed a significant association between severity of hypertension following aflibercept and survival improvement. CONCLUSIONS Aflibercept showed promising activity in patients with metastatic melanoma of cutaneous or uveal origin. Further evaluation of aflibercept as a single agent and in combination is warranted.
Collapse
|
103
|
|
104
|
Abstract
A large number of agents are currently in development for patients with metastatic melanoma. Potent small molecule inhibitors of mutant BRAF and the immunotherapy agent ipilimumab have demonstrated promising clinical activity and will likely change the standard of care in the future. However, only a fraction of patients currently receive durable benefit from immune therapies, and despite initial high response rates, resistance to the small molecule targeted agents eventually develops. Substantial opportunities exist for developing biomarkers that will lead to improved combinations and to choices for therapy after progression on a prior targeted agent. Development of clinically useful predictive biomarkers in tumor or blood has been particularly difficult for the immune therapies, owing to the complexity of interactions between tumor and host that impact on anti-tumor immune responses. In the setting of multiple active agents, and none capable of producing long-lasting benefit in most patients, it will be critical to develop assays to match individual patients with the treatment or treatments most likely to be effective to produce the greatest benefit in the overall population.
Collapse
|
105
|
Lutzky J. New therapeutic options in the medical management of advanced melanoma. ACTA ACUST UNITED AC 2011; 29:249-57. [PMID: 21277538 DOI: 10.1016/j.sder.2010.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
During the past 3 decades, the incidence, morbidity, and mortality of malignant melanoma have increased dramatically. Advanced melanoma has remained a disease that is for the most part incurable and has challenged all therapeutic efforts to make a dent in its natural history. Recent advances in the understanding of the molecular alterations in melanoma and in the immunologic mechanisms playing a role in this malignancy have brought hope that significant progress can be achieved, as evidenced by early encouraging clinical data. This review will summarize these recent developments and their impact on current clinical practice.
Collapse
Affiliation(s)
- Jose Lutzky
- Melanoma Program, Division of Hematology/Oncology, Mount Sinai Comprehensive Cancer Center, Miami Beach, FL, USA.
| |
Collapse
|
106
|
Abstract
Melanoma is the most aggressive form of skin cancer whose worldwide incidence is rising faster than any other cancer. Few treatment options are available to patients with metastatic disease, and standard chemotherapeutic agents are generally ineffective. Cytokines such as IFN-α or IL-2 can promote immune recognition of melanoma, occasionally inducing dramatic and durable clinical responses. Here, we discuss several immunomodulatory agents, the safety of which are being evaluated in clinical trials. Challenges include an incomplete understanding of signaling pathways, appropriate clinical dose and route, and systemic immunosuppression in advanced melanoma patients. We consider how targeted cytokine therapy will integrate into the clinical arena, as well as the low likelihood of success of single cytokine therapies. Evidence supports a synergy between cytokine immunotherapy and other therapeutic approaches in melanoma, and strengthening this area of research will improve our understanding of how to use cytokine therapy better.
Collapse
Affiliation(s)
- Courtney Nicholas
- The Ohio State University, Department of Internal Medicine, Division of Medical Oncology, Columbus, OH 43210, USA
| | - Gregory B Lesinski
- The Ohio State University, Department of Internal Medicine, Division of Medical Oncology, Columbus, OH 43210, USA
| |
Collapse
|
107
|
Abstract
The incidence of renal cell carcinoma (RCC) is increasing and outcomes remain poor. One-third of patients with localized disease will relapse, and 5-year survival for patients with metastatic disease is less than 10%. No molecular test is currently available to identify which patients who have undergone 'curative' surgery will relapse, and which patients will respond to targeted therapy. Some well characterized biochemical pathways, such as those associated with von Hippel-Lindau disease, are aberrantly regulated in RCC and are associated with histological subtype, but the understanding of these pathways contributes little to the clinical management of patients with RCC. Gene expression and sequencing studies have increased our understanding of the genetic basis of the disease but have failed to establish any unified classification to improve molecular stratification or to predict which patients are likely to relapse or respond to targeted therapy. Instead, they have served to highlight that RCC is heterogeneous at histological, morphological, and molecular levels, and that novel approaches are required to resolve the complexity of RCC prognostication and prediction of treatment response.
Collapse
|
108
|
Hiscox S, Barrett-Lee P, Nicholson RI. Therapeutic targeting of tumor-stroma interactions. Expert Opin Ther Targets 2011; 15:609-21. [PMID: 21388336 DOI: 10.1517/14728222.2011.561201] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Cancers exist within a complex microenvironment populated by diverse cell types within a protein-rich extracellular matrix. It is becoming increasingly apparent that molecular interactions between epithelial cells and cells in the surrounding stroma promote growth, invasion and spread of the tumor itself and thus represents a crucial underlying driving force in tumorigenesis. AREAS COVERED This article reviews how key interactions between tumor epithelial cells and surrounding mesenchymal and immune cells can promote tumor progression and highlights molecular elements that might represent novel therapeutic targets. EXPERT OPINION The tumor microenvironment is increasingly being viewed as a potential therapeutic target with a number of strategies being developed to disrupt tumor-stroma interactions, in order to delay or circumvent tumor progression. Targeting elements of the tumor microenvironment, or signaling pathways in tumor cells activated as a consequence of stromal interactions, may prove a useful therapeutic strategy to prevent tumor development and progression. However, given the tumor cells' ability to circumvent various therapeutic agents when given as monotherapy, the success of these agents is likely to be seen when used in combination with existing treatments.
Collapse
Affiliation(s)
- Stephen Hiscox
- Cardiff University, Welsh School of Pharmacy, Cardiff, UK.
| | | | | |
Collapse
|
109
|
Bracarda S, Sisani M, Del Buono S, Ishiwa O, Montagnani F. Biologic tools to personalize treatment in genitourinary cancers. Crit Rev Oncol Hematol 2011; 84 Suppl 1:e42-8. [PMID: 21232974 DOI: 10.1016/j.critrevonc.2010.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 08/24/2010] [Accepted: 09/16/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Genitourinary (GU) cancers are a major healthcare issue in modern oncology. In the last decade many efforts have been made to develop new treatment options but with the possible exception of renal cell carcinoma, very few steps ahead have been taken. At the same time, a wide variety of molecular markers, potentially helpful in identifying patient subpopulation most likely to benefit from a specific treatment have been identified. Our goal is to clarify if biomarkers could be used at present to personalize treatment for GU cancers. MATERIALS AND METHODS Literature was search using PubMed and EMBASE using different terms and combinations regarding possible prognostic and predictive markers in renal, prostate and urothelial cancers. RESULTS 3546 articles were retrieved. After excluding duplications, preclinical studies and factors without possible predictive value 654 publications remain. N-telopeptide, HER2/neu, EGFR, and p53 in prostate cancer, sVEGF-A for RCC and EMMPRIN and Survivin in urothelial cancer were among those identified. After a careful examination of published data, none of them reached a sufficient evidence to be suggested for use outside of clinical trials. CONCLUSIONS To date any reliable biomarkers has been validated for tailored treatments approaches in GU cancer. Future studies focusing on this issue are urgently needed.
Collapse
Affiliation(s)
- Sergio Bracarda
- U.O.C. Medical Oncology, Department of Oncology, San Donato Hospital, AUSL8 Arezzo, Italy.
| | | | | | | | | |
Collapse
|
110
|
Abstract
PURPOSE OF REVIEW To review the latest status on prognostic factors in renal cell carcinoma (RCC). RECENT FINDINGS Many predictive and prognostic factors can help differentiate between favorable and unfavorable RCC phenotypes. There currently exist several clinical and/or pathological, and biological factors, which have been exclusively tested and used in predictive and prognostic models. Nonetheless, the search for highly informative and reliable factors of disease characteristics and progression continues. CONCLUSION Over the last decade, an increase occurred in the number of models that can predict the treated natural history of RCC. Many of these novel models and previously developed models are tested in a head-to-head fashion, with the intent of identifying the most accurate and valuable tools for clinical practice. Novel prognostic factors and more up-to-date models are urgently needed for patients with metastatic RCC, especially in the era of targeted therapies. This should represent the focus of contemporary prognostic modeling in RCC.
Collapse
|
111
|
Del Vecchio M, Mortarini R, Canova S, Di Guardo L, Pimpinelli N, Sertoli MR, Bedognetti D, Queirolo P, Morosini P, Perrone T, Bajetta E, Anichini A. Bevacizumab plus fotemustine as first-line treatment in metastatic melanoma patients: clinical activity and modulation of angiogenesis and lymphangiogenesis factors. Clin Cancer Res 2010; 16:5862-72. [PMID: 21030496 DOI: 10.1158/1078-0432.ccr-10-2363] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess the clinical and biological activity of the association of bevacizumab and fotemustine as first-line treatment in advanced melanoma patients. EXPERIMENTAL DESIGN Previously untreated, metastatic melanoma patients (n = 20) received bevacizumab (at 15 mg/kg every 3 weeks) and fotemustine (100 mg/m² by intravenous administration on days 1, 8, and 15, repeated after 4 weeks) in a multicenter, single-arm, open-label, phase II study. Primary endpoint was the best overall response rate; other endpoints were toxicity, time to progression (TTP), and overall survival (OS). Serum cytokines, angiogenesis, and lymphangiogenesis factors were monitored by multiplex arrays and by in vitro angiogenesis assays. Effects of fotemustine on melanoma cells, in vitro, on vascular endothelial growth factor (VEGF)-C release and apoptosis were assessed by ELISA and flow cytometry, respectively. RESULTS One complete response, 2 partial responses (PR), and 10 patients with stable disease were observed. TTP and OS were 8.3 and 20.5 months, respectively. Fourteen patients experienced adverse events of toxicity grade 3-4. Serum VEGF-A levels in evaluated patients (n = 15) and overall serum proangiogenic activity were significantly inhibited. A significant reduction in VEGF-C levels was found in several post-versus pretherapy serum samples. In vitro, fotemustine inhibited VEGF-C release by melanoma cells without inducing significant cell death. Serum levels of interleukin (IL)-10 and IL-12p70 showed the highest levels in sera of PR patients, compared with patients with stable or progressive disease whereas IL-23 showed the opposite pattern. CONCLUSIONS The combination of bevacizumab plus fotemustine has clinical activity in advanced melanoma and promotes systemic modulation of angiogenesis and lymphangiogenesis factors.
Collapse
Affiliation(s)
- Michele Del Vecchio
- Unit of Medical Oncology 2, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Biomarker alterations with metronomic use of low-dose zoledronic acid for breast cancer patients with bone metastases and potential clinical significance. Breast Cancer Res Treat 2010; 124:733-43. [DOI: 10.1007/s10549-010-1183-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 09/16/2010] [Indexed: 11/26/2022]
|
113
|
Garcia JA, Mekhail T, Elson P, Triozzi P, Nemec C, Dreicer R, Bukowski RM, Rini BI. Clinical and immunomodulatory effects of bevacizumab and low-dose interleukin-2 in patients with metastatic renal cell carcinoma: results from a phase II trial. BJU Int 2010; 107:562-70. [PMID: 20840548 DOI: 10.1111/j.1464-410x.2010.09573.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Low-dose interleukin-2 (IL-2) is a historical treatment for metastatic renal cell carcinoma (mRCC). Increased vascular endothelial growth factor (VEGF) levels inhibit dendritic cell (DC) differentiation and augment production of immunosuppressive regulatory T (Treg) cells. Bevacizumab is an antibody that binds to VEGF, has activity in mRCC and may augment the anti-tumour immune effects of IL-2. To determine the clinical and immunomodulatory effects of this combination, a prospective, phase II trial of bevacizumab plus low-dose IL-2 was conducted. PATIENTS AND METHODS Patients with untreated mRCC received bevacizumab (10 mg/kg i.v. every 2 weeks) and IL-2 (125,000 units/kg/day subcutaneously from Monday to Friday for 6 consecutive weeks followed by a 2-week rest period). Endpoints included progression-free survival, Response Evaluation Criteria in Solid Tumors-defined objective response rate, immunomodulatory effects and safety. RESULTS Between January 2005 and September 2007, twenty-six patients with untreated mRCC were enrolled. The median progression-free survival was 9.6 months (95% CI, 4.1-16.9 months) The objective response rate was 15% and an additional 38% of patients had tumour burden reduction of <30%. Grade 3 constitutional adverse events (fatigue, fever/chills) and neutropenia were observed in 42% and 12% of patients, respectively. Peripheral blood CD1c(+) myeloid and CD303(+) plasmacytoid DC increased during treatment as did IL-8 levels and CD4(+) CD25(+) FoxP3(+) Treg cells. No changes in T helper type 1/2-associated cytokines were observed. CONCLUSION Bevacizumab plus low-dose IL-2 has modest clinical activity in mRCC. Toxicity was largely IL-2 related without enhancement of bevacizumab-related toxicity. Biological data indicate inhibition of VEGF levels and increase of immunosuppressive Treg cells without an effect on DC activation.
Collapse
Affiliation(s)
- Jorge A Garcia
- Department of Solid Tumor Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland Clinic Glickman Urological and Kidney Institute, Cleveland, OH, USA.
| | | | | | | | | | | | | | | |
Collapse
|
114
|
Ljungberg B, Cowan NC, Hanbury DC, Hora M, Kuczyk MA, Merseburger AS, Patard JJ, Mulders PF, Sinescu IC. EAU Guidelines on Renal Cell Carcinoma: The 2010 Update. Eur Urol 2010; 58:398-406. [PMID: 20633979 DOI: 10.1016/j.eururo.2010.06.032] [Citation(s) in RCA: 1039] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 06/21/2010] [Indexed: 12/22/2022]
|
115
|
Affiliation(s)
- Ryan J Sullivan
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
116
|
Current world literature. Curr Opin Urol 2010; 20:443-51. [PMID: 20679773 DOI: 10.1097/mou.0b013e32833dde0d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
117
|
Pérez-Gracia JL, Gúrpide A, Ruiz-Ilundain MG, Alfaro Alegría C, Colomer R, García-Foncillas J, Melero Bermejo I. Selection of extreme phenotypes: the role of clinical observation in translational research. Clin Transl Oncol 2010; 12:174-80. [PMID: 20231122 DOI: 10.1007/s12094-010-0487-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Systematic collection of phenotypes and their correlation with molecular data has been proposed as a useful method to advance in the study of disease. Although some databases for animal species are being developed, progress in humans is slow, probably due to the multifactorial origin of many human diseases and to the intricacy of accurately classifying phenotypes, among other factors. An alternative approach has been to identify and to study individuals or families with very characteristic, clinically relevant phenotypes. This strategy has shown increased efficiency to identify the molecular features underlying such phenotypes. While on most occasions the subjects selected for these studies presented harmful phenotypes, a few studies have been performed in individuals with very favourable phenotypes. The consistent results achieved suggest that it seems logical to further develop this strategy as a methodology to study human disease, including cancer. The identification and the study with high-throughput techniques of individuals showing a markedly decreased risk of developing cancer or of cancer patients presenting either an unusually favourable prognosis or striking responses following a specific treatment, might be promising ways to maximize the yield of this approach and to reveal the molecular causes that explain those phenotypes and thus highlight useful therapeutic targets. This manuscript reviews the current status of selection of extreme phenotypes in cancer research and provides directions for future development of this methodology.
Collapse
Affiliation(s)
- José Luis Pérez-Gracia
- Medical Oncology Department, Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain.
| | | | | | | | | | | | | |
Collapse
|
118
|
Alexandrescu DT, Ichim TE, Riordan NH, Marincola FM, Di Nardo A, Kabigting FD, Dasanu CA. Immunotherapy for melanoma: current status and perspectives. J Immunother 2010; 33:570-90. [PMID: 20551839 PMCID: PMC3517185 DOI: 10.1097/cji.0b013e3181e032e8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Immunotherapy is an important modality in the therapy of patients with malignant melanoma. As our knowledge about this disease continues to expand, so does the immunotherapeutic armamentarium. Nevertheless, successful preclinical models do not always translate into clinically meaningful results. The authors give a comprehensive analysis of most recent advances in the immune anti-melanoma therapy, including interleukins, interferons, other cytokines, adoptive immunotherapy, biochemotherapy, as well as the use of different vaccines. We also present the fundamental concepts behind various immune enhancement strategies, passive immunotherapy, as well as the use of immune adjuvants. This review brings into discussion the results of newer and older clinical trials, as well as potential limitations and drawbacks seen with the utilization of various immune therapies in malignant melanoma. Development of novel therapeutic approaches, along with optimization of existing therapies, continues to hold a great promise in the field of melanoma therapy research. Use of anti-CTLA4 and anti-PD1 antibodies, realization of the importance of co-stimulatory signals, which translated into the use of agonist CD40 monoclonal antibodies, as well as activation of innate immunity through enhanced expression of co-stimulatory molecules on the surface of dendritic cells by TLR agonists are only a few items on the list of recent advances in the treatment of melanoma. The need to engineer better immune interactions and to boost positive feedback loops appear crucial for the future of melanoma therapy, which ultimately resides in our understanding of the complexity of immune responses in this disease.
Collapse
Affiliation(s)
- Doru T Alexandrescu
- Division of Dermatology, University of California at San Diego, San Diego, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
119
|
Prognostic and predictive biomarkers in renal cell carcinoma. Target Oncol 2010; 5:85-94. [DOI: 10.1007/s11523-010-0143-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 06/17/2010] [Indexed: 11/27/2022]
|
120
|
Abstract
Biomarkers play an important role in the diagnosis and prognostic classification of various cancers and can be useful in monitoring the patient's clinical course of disease and response to therapy. Generally, biomarkers are proteins and their expressions are associated with malignant disease. In the majority of cases, the marker molecules are expressed by the tumour cells themselves or by the tumour microenvironment cells. Thus, most biomarkers can primarily be found in malignant tissues, but after active secretion or passive release at tumour destruction, they become detectable in body fluids such as blood. Besides morphological and histopathological biomarkers (anatomic site, type of the primary tumour, tumour size, invasion depth, vascular invasion and ulceration), an increasing variety of serological markers have been identified, providing the possibility of a more detailed diagnostic and prognostic subgrouping of tumour entities, up to and even changing existing classification systems. The goal of this review is to provide an overview of old and more recent serological biomarkers in malignant melanoma. We will first focus on confirmed and nonconfirmed serum tumour markers, followed by proteomic profiling, an innovative approach to identify new and better serological biomarkers in melanoma, and ending with the predictive factors for treatments in this pathology.
Collapse
|
121
|
Bedognetti D, Wang E, Sertoli MR, Marincola FM. Gene-expression profiling in vaccine therapy and immunotherapy for cancer. Expert Rev Vaccines 2010; 9:555-65. [PMID: 20518712 PMCID: PMC3411321 DOI: 10.1586/erv.10.55] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The identification of tumor antigens recognized by T cells led to the design of therapeutic strategies aimed at eliciting adaptive immune responses. The last decade of experience has shown that, although active immunization can induce enhancement of anticancer T-cell precursors (easily detectable in standard assays), most often they are unable to induce tumor regression and, consequently, have scarcely any impact on overall survival. Moreover, in the few occasions when tumor rejection occurs, the mechanisms determining this phenomenon remain poorly understood, and data derived from in vivo human observations are rare. The advent of high-throughput gene-expression analysis (microarrays) has cast new light on unrecognized mechanisms that are now deemed to be central for the development of efficient immune-mediated tumor rejection. The aim of this article is to review the data on the molecular signature associated with this process. We believe that the description of how the mechanism of immune-mediated tissue destruction occurs would contribute to our understanding of why it happens, thereby allowing us to develop more effective immune therapeutic strategies.
Collapse
Affiliation(s)
- Davide Bedognetti
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
- S.C. Oncologia Medica B, Department of Medical Oncology, National Cancer Research Institute, Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Oncology, Biology and Genetics, University of Genoa, Genoa, Italy
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario Roberto Sertoli
- S.C. Oncologia Medica B, Department of Medical Oncology, National Cancer Research Institute, Genoa, Italy
- Department of Oncology, Biology and Genetics, University of Genoa, Genoa, Italy
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
122
|
|
123
|
Kao J, Ko EC, Eisenstein S, Sikora AG, Fu S, Chen SH. Targeting immune suppressing myeloid-derived suppressor cells in oncology. Crit Rev Oncol Hematol 2010; 77:12-9. [PMID: 20304669 DOI: 10.1016/j.critrevonc.2010.02.004] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 01/10/2010] [Accepted: 02/05/2010] [Indexed: 12/11/2022] Open
Abstract
Emerging data suggests that host immune cells with a suppressive phenotype represent a significant hurdle to successful therapy for metastatic cancer. Among the suppressor cells, T regulatory cells (Treg) and myeloid-derived suppressor cells (MDSC) are significantly increased in hosts with advanced malignancies. MDSC mediate the suppression of the tumor antigen-specific T cell response through the induction of T cell anergy and the development of Treg in tumor-bearing mice. These results provide robust evidence of an in vivo immunoregulatory function of MDSC in the establishment of tumor antigen-specific tolerance and the development of Treg in tumor-bearing hosts. To achieve effective anti-tumor immunity, tumor-induced immunosuppression must be reversed. Our preliminary results indicate that c-kit ligand (stem cell factor) expressed by tumor cells may be required for MDSC accumulation in tumor-bearing mice, and that blocking the c-kit ligand/c-kit receptor interaction can prevent the development of Treg and reverse immune tolerance induced by MDSC. Since c-kit can be readily inhibited by several small molecule inhibitors including imatinib, sunitinib and dasatinib, targeting immune suppressing cells can be readily accomplished in the clinic.
Collapse
Affiliation(s)
- Johnny Kao
- Department of Radiation Oncology, Mount Sinai School of Medicine, New York, NY 10029, United States.
| | | | | | | | | | | |
Collapse
|
124
|
Ribatti D, Annese T, Longo V. Angiogenesis and melanoma. Cancers (Basel) 2010; 2:114-32. [PMID: 24281035 PMCID: PMC3827594 DOI: 10.3390/cancers2010114] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 02/10/2010] [Accepted: 02/24/2010] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis occurs in pathological conditions, such as tumors, where a specific critical point in tumor progression is the transition from the avascular to the vascular phase. Tumor angiogenesis depends mainly on the release by neoplastic cells of growth factors specific for endothelial cells, which are able to stimulate the growth of the host's blood vessels. This article summarizes the literature concerning the relationship between angiogenesis and human melanoma progression. The recent applications of antiangiogenic agents which interfere with melanoma progression are also described.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Human Anatomy and Histology, University of Bari Medical School, Piazza G. Cesare, 11, Policlinico 70124, Bari, Italy.
| | | | | |
Collapse
|
125
|
Hamilton HK, Rose AE, Christos PJ, Shapiro RL, Berman RS, Mazumdar M, Ma MW, Krich D, Liebes L, Brooks PC, Osman I. Increased shedding of HU177 correlates with worse prognosis in primary melanoma. J Transl Med 2010; 8:19. [PMID: 20178639 PMCID: PMC2837640 DOI: 10.1186/1479-5876-8-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 02/23/2010] [Indexed: 01/07/2023] Open
Abstract
Background Increased levels of cryptic collagen epitope HU177 in the sera of melanoma patients have been shown to be associated with thicker primary melanomas and with the nodular histologic subtype. In this study, we investigate the association between HU177 shedding in the sera and clinical outcome in terms of disease-free survival (DFS) and overall survival (OS). Methods Serum samples from 209 patients with primary melanoma prospectively enrolled in the Interdisciplinary Melanoma Cooperative Group at the New York University Langone Medical Center (mean age = 58, mean thickness = 2.09 mm, stage I = 136, stage II = 41, stage III = 32, median follow-up = 54.9 months) were analyzed for HU177 concentration using a validated ELISA assay. HU177 serum levels at the time of diagnosis were used to divide the study cohort into two groups: low and high HU177. DFS and OS were estimated by Kaplan-Meier survival analysis, and the log-rank test was used to compare DFS and OS between the two HU177 groups. Multivariate Cox proportional hazards regression models were employed to examine the independent effect of HU177 category on DFS and OS. Results HU177 sera concentrations ranged from 0-139.8 ng/ml (mean and median of 6.2 ng/ml and 3.7 ng/ml, respectively). Thirty-eight of the 209 (18%) patients developed recurrences, and 34 of the 209 (16%) patients died during follow-up. Higher HU177 serum level was associated with an increased rate of melanoma recurrence (p = 0.04) and with increasing mortality (p = 0.01). The association with overall survival remained statistically significant after controlling for thickness and histologic subtype in a multivariate model (p = 0.035). Conclusions Increased shedding of HU177 in the serum of primary melanoma patients is associated with poor prognosis. Further studies are warranted to determine the clinical utility of HU177 in risk stratification compared to the current standard of care.
Collapse
Affiliation(s)
- Heather K Hamilton
- Department of Dermatology, New York University School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Monsurrò V, Beghelli S, Wang R, Barbi S, Coin S, Di Pasquale G, Bersani S, Castellucci M, Sorio C, Eleuteri S, Worschech A, Chiorini JA, Pederzoli P, Alter H, Marincola FM, Scarpa A. Anti-viral state segregates two molecular phenotypes of pancreatic adenocarcinoma: potential relevance for adenoviral gene therapy. J Transl Med 2010; 8:10. [PMID: 20113473 PMCID: PMC2845551 DOI: 10.1186/1479-5876-8-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 01/29/2010] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) remains a leading cause of cancer mortality for which novel gene therapy approaches relying on tumor-tropic adenoviruses are being tested. Methods We obtained the global transcriptional profiling of primary PDAC using RNA from eight xenografted primary PDAC, three primary PDAC bulk tissues, three chronic pancreatitis and three normal pancreatic tissues. The Affymetrix GeneChip HG-U133A was used. The results of the expression profiles were validated applying immunohistochemical and western blot analysis on a set of 34 primary PDAC and 10 established PDAC cell lines. Permissivity to viral vectors used for gene therapy, Adenovirus 5 and Adeno-Associated Viruses 5 and 6, was assessed on PDAC cell lines. Results The analysis of the expression profiles allowed the identification of two clearly distinguishable phenotypes according to the expression of interferon-stimulated genes. The two phenotypes could be readily recognized by immunohistochemical detection of the Myxovirus-resistance A protein, whose expression reflects the activation of interferon dependent pathways. The two molecular phenotypes discovered in primary carcinomas were also observed among established pancreatic adenocarcinoma cell lines, suggesting that these phenotypes are an intrinsic characteristic of cancer cells independent of their interaction with the host's microenvironment. The two pancreatic cancer phenotypes are characterized by different permissivity to viral vectors used for gene therapy, as cell lines expressing interferon stimulated genes resisted to Adenovirus 5 mediated lysis in vitro. Similar results were observed when cells were transduced with Adeno-Associated Viruses 5 and 6. Conclusion Our study identified two molecular phenotypes of pancreatic cancer, characterized by a differential expression of interferon-stimulated genes and easily recognized by the expression of the Myxovirus-resistance A protein. We suggest that the detection of these two phenotypes might help the selection of patients enrolled in virally-mediated gene therapy trials.
Collapse
Affiliation(s)
- Vladia Monsurrò
- Department of Pathology, University of Verona Medical School, Verona, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Chavez ARDV, Buchser W, Basse PH, Liang X, Appleman LJ, Maranchie JK, Zeh H, de Vera ME, Lotze MT. Pharmacologic administration of interleukin-2. Ann N Y Acad Sci 2010; 1182:14-27. [PMID: 20074271 DOI: 10.1111/j.1749-6632.2009.05160.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The development of biologic therapies for patients with cancer has in part been impeded by the extraordinary complexity and intrinsic feedback mechanisms promoting homeostasis in tissue injury, repair, inflammation, and immunity. Recombinant interleukin 2 (IL-2) therapy was initiated in 1984 based on its role as the prototypic T-cell growth factor, with novel roles deduced late after its FDA approval in regulating not only effector T cells but also regulatory T cells. Complicating its application, even in the most sophisticated centers, has been the manageable but difficult toxicities attendant on its use in spite of clear evidence of complete responses in 5-10% of treated patients with melanoma and renal cell carcinoma with extraordinary durability lasting now for almost 25 years, thus tantamount to "cures." Although efforts have been made to diminish toxicity or enhance efficacy the only substantive advance in combination therapy has been the application of tumor-infiltrating lymphocytes and the antibody to CTLA4. A deeper understanding of the "limiting" toxicity associated with mild flu-like symptoms and more debilitating cytokine "storm" not forthcoming. Here we propose the notion that the systemic syndrome associated with IL-2 administration is due to global cytokine-induced autophagy and temporally limited tissue dysfunction. The possible role of autophagy inhibitors to enhance efficacy and limit toxicity as well as possible problems with this approach are considered.
Collapse
|
128
|
Donnelly RP, Young HA, Rosenberg AS. An overview of cytokines and cytokine antagonists as therapeutic agents. Ann N Y Acad Sci 2010; 1182:1-13. [PMID: 20074270 DOI: 10.1111/j.1749-6632.2009.05382.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cytokine-based therapies have the potential to provide novel treatments for cancer, autoimmune diseases, and many types of infectious disease. However, to date, the full clinical potential of cytokines as drugs has been limited by a number of factors. To discuss these limitations and explore ways to overcome them, the FDA partnered with the New York Academy of Sciences in March 2009 to host a two-day forum to discuss more effective ways to harness the clinical potential of cytokines and cytokine antagonists as therapeutic agents. The first day was focused primarily on the use of recombinant cytokines as therapeutic agents for treatment of human diseases. The second day focused largely on the use of cytokine antagonists as therapeutic agents for treatment of human diseases. This issue of the Annals includes more than a dozen papers that summarize much of the information that was presented during this very informative two-day conference.
Collapse
Affiliation(s)
- Raymond P Donnelly
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA.
| | | | | |
Collapse
|
129
|
Mehnert JM, McCarthy MM, Jilaveanu L, Flaherty KT, Aziz S, Camp RL, Rimm DL, Kluger HM. Quantitative expression of VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 in melanoma tissue microarrays. Hum Pathol 2009; 41:375-84. [PMID: 20004943 DOI: 10.1016/j.humpath.2009.08.016] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Revised: 08/10/2009] [Accepted: 08/18/2009] [Indexed: 12/19/2022]
Abstract
Angiogenesis is required for progression and metastasis of melanoma. Analysis of angiogenic molecules in benign and malignant tissues may allow identification of markers useful for prediction of sensitivity to antiangiogenic agents. We hypothesized that differential expression of vascular endothelial growth factor (VEGF) and its receptors VEGF-R1, VEGF-R2, and VEGF-R3 would be higher in melanomas than nevi and higher in advanced melanoma. Using automated quantitative analysis, we quantified VEGF, -R1, -R2 and -R3 expression in melanoma tissue microarrays composed of 540 nevi and 468 melanoma specimens (198 primaries, 270 metastases). VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 expression was significantly higher in melanomas than nevi by unpaired t tests (P < .0001). VEGF-R2 expression was higher in metastatic specimens (P < .0001), but VEGF-R3 expression was higher in primaries (P < .0001). VEGF was coexpressed with all 3 receptors when assessed by Spearman's rank correlation. VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 expression is higher in melanomas than nevi. Higher expression of VEGF-R2 was found in metastases versus primaries, supporting the idea that selection for an angiogenic phenotype in metastatic melanoma is conferred via up-regulation of VEGF-R2. However, higher expression of VEGF-R3 was seen on primary lesions, potentially implicating this receptor in initiation of lymphatic tumor spread. Clinical trials using antiangiogenic agents in melanoma should include correlative assays of VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 as biomarkers of response to therapy, preferably using quantitative methods such as automated quantitative analysis. Such assessments could assist with evaluation of these molecules as therapeutic targets in melanoma, ultimately facilitating improved selection of patients for treatment.
Collapse
Affiliation(s)
- Janice M Mehnert
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | | | | | |
Collapse
|
130
|
Guminski AD, Thompson JF. Predicting response to IL-2 therapy for metastatic melanoma. Expert Rev Anticancer Ther 2009; 9:1571-5. [PMID: 19895241 DOI: 10.1586/era.09.131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
131
|
Kotlan B, Stroncek DF, Marincola FM. Turning laboratory findings into therapy: a marathon goal that has to be reached. Pol Arch Intern Med 2009; 119:586-94. [PMID: 19776705 PMCID: PMC3528796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The mission of translational research involves difficult tasks to be accomplished for its ultimate goal, i.e., the introduction of novel, effective therapeutic strategies in the clinic to diminish human suffering and cure life-threatening diseases. Translational research (also referred to as translational medicine) facilitates the translation of investment in biomedical research into successful medical treatment. This includes the transfer of diagnostic and therapeutic advances by proving their efficacy in large evidence-based trials. Through the study of humans novel insights about disease are brought back to the laboratory to identify new, observation-based strategies. This "two-way road" ("bench to bedside and bedside to bench") process includes formulating guidelines for drug development and principles for new therapeutic strategies; initiating clinical investigations that provide the biological basis for new therapies, and related clinical trials; defining therapeutic targets and clinical endpoints. It requires a systematic approach beginning with specimen sampling, patient data collection, laboratory investigations, data analysis, preclinical testing, clinical trials, treatment efficacy monitoring, and finally the evaluation of therapeutic result. The marathon well symbolizes the enormous efforts undertaken by clinicians, scientists, regulators, ethicists, patient advocates, drug developers, and others, coordinately attempting to overcome obstacles along this road toward the final "marathon goal in medicine".
Collapse
Affiliation(s)
- Beatrix Kotlan
- Center of Surgical and Molecular Tumor Pathology, National Institute of Oncology, Budapest, Hungary
| | - David F. Stroncek
- Chief Cell Processing Section, Department of Transfusion Medicine, National Institute of Health, Bethesda, MD, United States
| | - Francesco M. Marincola
- Chief Infectious Disease and Immunogenetics Section, Department of Transfusion Medicine, Center for Human Immunology, National Institute of Health, Bethesda, MD, United States
| |
Collapse
|
132
|
Tahara H, Sato M, Thurin M, Wang E, Butterfield LH, Disis ML, Fox BA, Lee PP, Khleif SN, Wigginton JM, Ambs S, Akutsu Y, Chaussabel D, Doki Y, Eremin O, Fridman WH, Hirohashi Y, Imai K, Jacobson J, Jinushi M, Kanamoto A, Kashani-Sabet M, Kato K, Kawakami Y, Kirkwood JM, Kleen TO, Lehmann PV, Liotta L, Lotze MT, Maio M, Malyguine A, Masucci G, Matsubara H, Mayrand-Chung S, Nakamura K, Nishikawa H, Palucka AK, Petricoin EF, Pos Z, Ribas A, Rivoltini L, Sato N, Shiku H, Slingluff CL, Streicher H, Stroncek DF, Takeuchi H, Toyota M, Wada H, Wu X, Wulfkuhle J, Yaguchi T, Zeskind B, Zhao Y, Zocca MB, Marincola FM. Emerging concepts in biomarker discovery; the US-Japan Workshop on Immunological Molecular Markers in Oncology. J Transl Med 2009; 7:45. [PMID: 19534815 PMCID: PMC2724494 DOI: 10.1186/1479-5876-7-45] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 06/17/2009] [Indexed: 02/08/2023] Open
Abstract
Supported by the Office of International Affairs, National Cancer Institute (NCI), the "US-Japan Workshop on Immunological Biomarkers in Oncology" was held in March 2009. The workshop was related to a task force launched by the International Society for the Biological Therapy of Cancer (iSBTc) and the United States Food and Drug Administration (FDA) to identify strategies for biomarker discovery and validation in the field of biotherapy. The effort will culminate on October 28th 2009 in the "iSBTc-FDA-NCI Workshop on Prognostic and Predictive Immunologic Biomarkers in Cancer", which will be held in Washington DC in association with the Annual Meeting. The purposes of the US-Japan workshop were a) to discuss novel approaches to enhance the discovery of predictive and/or prognostic markers in cancer immunotherapy; b) to define the state of the science in biomarker discovery and validation. The participation of Japanese and US scientists provided the opportunity to identify shared or discordant themes across the distinct immune genetic background and the diverse prevalence of disease between the two Nations. Converging concepts were identified: enhanced knowledge of interferon-related pathways was found to be central to the understanding of immune-mediated tissue-specific destruction (TSD) of which tumor rejection is a representative facet. Although the expression of interferon-stimulated genes (ISGs) likely mediates the inflammatory process leading to tumor rejection, it is insufficient by itself and the associated mechanisms need to be identified. It is likely that adaptive immune responses play a broader role in tumor rejection than those strictly related to their antigen-specificity; likely, their primary role is to trigger an acute and tissue-specific inflammatory response at the tumor site that leads to rejection upon recruitment of additional innate and adaptive immune mechanisms. Other candidate systemic and/or tissue-specific biomarkers were recognized that might be added to the list of known entities applicable in immunotherapy trials. The need for a systematic approach to biomarker discovery that takes advantage of powerful high-throughput technologies was recognized; it was clear from the current state of the science that immunotherapy is still in a discovery phase and only a few of the current biomarkers warrant extensive validation. It was, finally, clear that, while current technologies have almost limitless potential, inadequate study design, limited standardization and cross-validation among laboratories and suboptimal comparability of data remain major road blocks. The institution of an interactive consortium for high throughput molecular monitoring of clinical trials with voluntary participation might provide cost-effective solutions.
Collapse
Affiliation(s)
- Hideaki Tahara
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Marimo Sato
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Magdalena Thurin
- Cancer Diagnosis Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, Maryland, 20852, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, Maryland, 20892, USA
| | - Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington, 98195, USA
| | - Bernard A Fox
- Earle A Chiles Research Institute, Robert W Franz Research Center, Providence Portland Medical Center, and Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, 97213, USA
| | - Peter P Lee
- Department of Medicine, Division of Hematology, Stanford University, Stanford, California, 94305, USA
| | - Samir N Khleif
- Cancer Vaccine Section, NCI, NIH, Bethesda, Maryland, 20892, USA
| | - Jon M Wigginton
- Discovery Medicine-Oncology, Bristol-Myers Squibb Inc., Princeton, New Jersey, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center of Cancer Research, NCI, NIH, Bethesda, Maryland, 20892, USA
| | - Yasunori Akutsu
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Damien Chaussabel
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - Yuichiro Doki
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Oleg Eremin
- Section of Surgery, Biomedical Research Unit, Nottingham Digestive Disease Centre, University of Nottingham, NG7 2UH, UK
| | - Wolf Hervé Fridman
- Centre de la Reserche des Cordeliers, INSERM, Paris Descarte University, 75270 Paris, France
| | | | - Kohzoh Imai
- Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - James Jacobson
- Cancer Diagnosis Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, Maryland, 20852, USA
| | - Masahisa Jinushi
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akira Kanamoto
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | - Kazunori Kato
- Department of Molecular Medicine, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - John M Kirkwood
- Departments of Medicine, Surgery and Immunology, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Thomas O Kleen
- Cellular Technology Ltd, Shaker Heights, Ohio, 44122, USA
| | - Paul V Lehmann
- Cellular Technology Ltd, Shaker Heights, Ohio, 44122, USA
| | - Lance Liotta
- Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, 10900, USA
| | - Michael T Lotze
- Illman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, USA
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department. of Oncology, University, Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, IRCCS, Aviano, 53100, Italy
| | - Anatoli Malyguine
- Laboratory of Cell Mediated Immunity, SAIC-Frederick, Inc. NCI-Frederick, Frederick, Maryland, 21702, USA
| | - Giuseppe Masucci
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, 171 76, Sweden
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shawmarie Mayrand-Chung
- The Biomarkers Consortium (BC), Public-Private Partnership Program, Office of the Director, NIH, Bethesda, Maryland, 20892, USA
| | - Kiminori Nakamura
- Department of Molecular Medicine, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Hiroyoshi Nishikawa
- Department of Cancer Vaccine, Department of Immuno-gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - A Karolina Palucka
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - Emanuel F Petricoin
- Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, 10900, USA
| | - Zoltan Pos
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, Maryland, 20892, USA
| | - Antoni Ribas
- Department of Medicine, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, 90095, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Foundation, Istituto Nazionale Tumori, Milan, 20100, Italy
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Shiku
- Department of Cancer Vaccine, Department of Immuno-gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Craig L Slingluff
- Department of Surgery, Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908, USA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, DCTD, NCI, NIH, Rockville, Maryland, 20892, USA
| | - David F Stroncek
- Cell Therapy Section (CTS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland, 20892, USA
| | - Hiroya Takeuchi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Toyota
- Department of Biochemistry, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Hisashi Wada
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Xifeng Wu
- Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Julia Wulfkuhle
- Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, 10900, USA
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | | | - Yingdong Zhao
- Biometric Research Branch, NCI, NIH, Bethesda, Maryland, 20892, USA
| | | | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, Maryland, 20892, USA
| |
Collapse
|
133
|
Kirkwood JM, Tarhini AA. Biomarkers of Therapeutic Response in Melanoma and Renal Cell Carcinoma: Potential Inroads to Improved Immunotherapy. J Clin Oncol 2009; 27:2583-5. [DOI: 10.1200/jco.2008.21.1540] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- John M. Kirkwood
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Ahmad A. Tarhini
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| |
Collapse
|
134
|
Wang E, Monaco A, Monsurró V, Sabatino M, Pos Z, Uccellini L, Wang J, Worschech A, Stroncek DF, Marincola FM. Antitumor vaccines, immunotherapy and the immunological constant of rejection. IDRUGS : THE INVESTIGATIONAL DRUGS JOURNAL 2009; 12:297-301. [PMID: 19431094 PMCID: PMC3410731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Anticancer vaccines have not matched the clinical expectations projected from their ability to induce consistently systemic anticancer T-cell responses. Thus, a dichotomy is observed between the immunological and clinical endpoints of anticancer immunization. Anticancer vaccines have clearly demonstrated that highly specific T-cell responses can be induced that can recognize autologous cancer antigens in patients with cancer. This ability is an outstanding achievement of modern biotechnology, yielding one of the most specific types of potential anticancer reagents. However, systemic, vaccine-induced anticancer responses exemplify a broader immunological paradox: cytotoxic T-cells can coexist within the same organism with their target cells not only in the context of cancer, but also in the context of chronic infections, well-controlled allo-transplant reactions and autoimmunity. According to this view, anticancer immune responses are a facet of a tissue-specific autoimmune phenomenon specific for cancer tissue that may or may not result in the successful immune-destruction of target cells, depending on an assortment of genetic factors related to the background of the host or evolving phenotypes of a heterogeneous cancer environment. This feature article summarizes the current understanding of the mechanisms leading to tumor rejection in humans as well as in experimental models, in the context of the broader immunological phenomenon leading to tissue-specific destruction. Anticancer vaccines that may not induce clinically significant anticancer responses independently could function as a unique tool to enhance the specificity of the response of the host against cancer, provided that strategies are implemented to amplify the immune reaction initiated by vaccine-induced antibodies and/or T-cells.
Collapse
Affiliation(s)
- Ena Wang
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Alessandro Monaco
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Vladia Monsurró
- University of Verona Medical School, Department of Pathology, c/o Policlinico GB Rossi, P le LA Scuro 10, 37134, Verona, Italy
| | - Marianna Sabatino
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Zoltan Pos
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Lorenzo Uccellini
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Jeanne Wang
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Andrea Worschech
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
- Genelux Corp, San Diego Science Center, 3030 Bunker Hill Street, Suite 310, San Diego, CA 92109, USA
| | - David F Stroncek
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| | - Francesco M Marincola
- National Institutes of Health, Department of Transfusion Medicine, Building 10, Room 1C711, Clinical Center, Bethesda, MD 20892, USA
| |
Collapse
|
135
|
Butterfield LH, Disis ML, Fox BA, Lee PP, Khleif SN, Thurin M, Trinchieri G, Wang E, Wigginton J, Chaussabel D, Coukos G, Dhodapkar M, Håkansson L, Janetzki S, Kleen TO, Kirkwood JM, Maccalli C, Maecker H, Maio M, Malyguine A, Masucci G, Palucka AK, Potter DM, Ribas A, Rivoltini L, Schendel D, Seliger B, Selvan S, Slingluff CL, Stroncek DF, Streicher H, Wu X, Zeskind B, Zhao Y, Zocca MB, Zwierzina H, Marincola FM. A systematic approach to biomarker discovery; preamble to "the iSBTc-FDA taskforce on immunotherapy biomarkers". J Transl Med 2008; 6:81. [PMID: 19105846 PMCID: PMC2630944 DOI: 10.1186/1479-5876-6-81] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 12/23/2008] [Indexed: 12/23/2022] Open
Abstract
The International Society for the Biological Therapy of Cancer (iSBTc) has initiated in collaboration with the United States Food and Drug Administration (FDA) a programmatic look at innovative avenues for the identification of relevant parameters to assist clinical and basic scientists who study the natural course of host/tumor interactions or their response to immune manipulation. The task force has two primary goals: 1) identify best practices of standardized and validated immune monitoring procedures and assays to promote inter-trial comparisons and 2) develop strategies for the identification of novel biomarkers that may enhance our understating of principles governing human cancer immune biology and, consequently, implement their clinical application. Two working groups were created that will report the developed best practices at an NCI/FDA/iSBTc sponsored workshop tied to the annual meeting of the iSBTc to be held in Washington DC in the Fall of 2009. This foreword provides an overview of the task force and invites feedback from readers that might be incorporated in the discussions and in the final document.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington, 98195, USA
| | - Bernard A Fox
- Earle A Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon, 97213, USA
- Department of Molecular Biology, OHSU Cancer Institute, Oregon Health and Science University, Portland, Oregon, 97213, USA
| | - Peter P Lee
- Department of Medicine, Division of Hematology, Stanford University, Stanford, California, 94305, USA
| | - Samir N Khleif
- Cancer Vaccine Section, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Magdalena Thurin
- Cancer Diagnosis Program, NCI, NIH, Rockville, Maryland, 20852, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, NCI, NIH, Frederick, Maryland, 21702, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology, National Institutes of Health, Bethesda, MD, USA
| | - Jon Wigginton
- Bristol Myers-Squibb, Princeton, New Jersey, 08540, USA
| | - Damien Chaussabel
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - George Coukos
- Center for Research on the Early Detection and Cure of Ovarian Cancer, University of Pennsylvania, Philadelphia 19104, USA
| | - Madhav Dhodapkar
- Department of Hematology, Yale University, New Haven, Connecticut 06510, USA
| | - Leif Håkansson
- Division of Clinical Tumor Immunology, University of Lund, 581 85, Sweden
| | | | - Thomas O Kleen
- Cellular Technology Limited, Shaker Heights, Ohio, 44122, USA
| | - John M Kirkwood
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Cristina Maccalli
- Unit of Immuno-Biotherapy of Solid Tumors, Department of Molecular Oncology, San Raffaele Scientific Institute DIBIT, Milan, 20132, Italy
| | - Holden Maecker
- Baylor Institute for Immunology Research, Dallas, 75204, Texas, USA
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department. of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, IRCCS, Aviano, 53100, Italy
| | - Anatoli Malyguine
- Laboratory of Cell Mediated Immunity, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
| | - Giuseppe Masucci
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, 171 76, Sweden
| | - A Karolina Palucka
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - Douglas M Potter
- Biostatistics Department, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, USA
| | - Antoni Ribas
- Department of Medicine, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, 90095, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Foundation, Istituto Nazionale Tumori, Milan, 20100, Italy
| | - Dolores Schendel
- Institute of Molecular Immunology, and Clinical Cooperation Group "Immune Monitoring" Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 81377, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther University, Halle Wittenberg, Halle (Saale), 06112, Germany
| | | | - Craig L Slingluff
- Department of Surgery, Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908, USA
| | - David F Stroncek
- Cell Therapy Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland, 20892, USA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, NCI, Bethesda, Maryland, 20852 USA
| | - Xifeng Wu
- Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | | | - Yingdong Zhao
- Biometrics Research Branch, NCI, NIH, Bethesda, Maryland, 20852, USA
| | | | - Heinz Zwierzina
- Department of Internal Medicine, Innsbruck Medical University, Innsbruck, 6020, Austria
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|