1
|
Tu SM, Trikannad AK, Vellanki S, Hussain M, Malik N, Singh SR, Jillella A, Obulareddy S, Malapati S, Bhatti SA, Arnaoutakis K, Atiq OT. Stem Cell Origin of Cancer: Clinical Implications beyond Immunotherapy for Drug versus Therapy Development in Cancer Care. Cancers (Basel) 2024; 16:1151. [PMID: 38539487 PMCID: PMC10969562 DOI: 10.3390/cancers16061151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 11/11/2024] Open
Abstract
Although immunotherapy has revolutionized cancer care, there is still an urgent need to enhance its efficacy and ensure its safety. A correct cancer theory and proper scientific method empower pertinent cancer research and enable effective and efficient drug versus therapy development for patient care. In this perspective, we revisit the concept of immune privilege in a cancer cell versus normal cell, as well as in a cancer stem cell versus normal stem cell. We re-examine whether effective immunotherapies are efficacious due to their anti-cancer and/or immune modulatory mechanisms. We reassess why checkpoint inhibitors (CPIs) are not equal. We reconsider whether one can attribute the utility of immunotherapy to specific cancer subtypes and its futility to certain tumor/immune compartments, components, and microenvironments. We propose ways and means to advance immunotherapy beyond CPIs by combining anti-PD1/L1 with various other treatment modalities according to an appropriate scientific theory, e.g., stem cell origin of cancer, and based on available clinical evidence, e.g., randomized clinical trials. We predict that a stem cell theory of cancer will facilitate the design of better and safer immunotherapy with improved selection of its use for the right patient with the right cancer type at the right time to optimize clinical benefits and minimize potential toxic effects and complications.
Collapse
Affiliation(s)
- Shi-Ming Tu
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA (M.H.); (N.M.); (S.R.S.); (A.J.); (S.O.); (S.M.); (S.A.B.); (K.A.); (O.T.A.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Rubanov A, Berico P, Hernando E. Epigenetic Mechanisms Underlying Melanoma Resistance to Immune and Targeted Therapies. Cancers (Basel) 2022; 14:cancers14235858. [PMID: 36497341 PMCID: PMC9738385 DOI: 10.3390/cancers14235858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is an aggressive skin cancer reliant on early detection for high likelihood of successful treatment. Solar UV exposure transforms melanocytes into highly mutated tumor cells that metastasize to the liver, lungs, and brain. Even upon resection of the primary tumor, almost thirty percent of patients succumb to melanoma within twenty years. Identification of key melanoma genetic drivers led to the development of pharmacological BRAFV600E and MEK inhibitors, significantly improving metastatic patient outcomes over traditional cytotoxic chemotherapy or pioneering IFN-α and IL-2 immune therapies. Checkpoint blockade inhibitors releasing the immunosuppressive effects of CTLA-4 or PD-1 proved to be even more effective and are the standard first-line treatment. Despite these major improvements, durable responses to immunotherapy and targeted therapy have been hindered by intrinsic or acquired resistance. In addition to gained or selected genetic alterations, cellular plasticity conferred by epigenetic reprogramming is emerging as a driver of therapy resistance. Epigenetic regulation of chromatin accessibility drives gene expression and establishes distinct transcriptional cell states. Here we review how aberrant chromatin, transcriptional, and epigenetic regulation contribute to therapy resistance and discuss how targeting these programs sensitizes melanoma cells to immune and targeted therapies.
Collapse
Affiliation(s)
- Andrey Rubanov
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Pietro Berico
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
3
|
Deboever N, Feldman HA, Hofstetter WL, Mehran RJ, Rajaram R, Rice DC, Roth JA, Sepesi B, Swisher SG, Vaporciyan AA, Walsh GL, Antonoff MB. The Role of Surgery in the Treatment of Melanoma Pulmonary Metastases in the Modern Era. J Surg Res 2022; 277:125-130. [PMID: 35489217 DOI: 10.1016/j.jss.2022.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/09/2022] [Accepted: 04/08/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The lung represents a frequent site of spread for metastatic melanoma, which has historically been managed with surgical resection achieving promising outcomes. We hypothesized that the role of surgery in the management of melanoma pulmonary metastases (MPM) is evolving among the development of less invasive diagnostic and novel systemic therapeutic strategies. MATERIALS AND METHODS A single-center thoracic surgery database was reviewed and patients who underwent surgical resection of MPM between 1998 and 2019 were identified. Demographic, clinicopathologic, and surgical data were collected and analyzed, as were the annual volumes and indications for surgical resection. A Cochran-Armitage test was used to assess the trend in surgical indication. RESULTS Three hundred and seventy seven surgical procedures for MPM were performed during the years of study in the care of 347 patients. Patients were predominantly male, with a mean age of 59.3 y. The mean number of annual resections was 17 and while this number initially increased from six in 1998 to a peak of 39 cases in 2008, a decline was subsequently observed. Diagnostic resection decreased from 22% in 1998-1999 to 5% at the peak of procedures in 2008-2009 and to 0 in 2018-2019 (P = 0.02). Curative resection increased from 44% in 1998-1999 to 73% in 2008-2009 (P < 0.001) and remained the dominant reason for surgery in later years. CONCLUSIONS Surgical indications in the management of MPM have transformed in conjunction with systemic modalities, and the volume of resections has decreased in the modern era. Despite innovations in systemic management and shifting goals of operative interventions, surgeons continue to play a vital role in caring for these patients with an advanced disease.
Collapse
Affiliation(s)
- Nathaniel Deboever
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hope A Feldman
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Reza J Mehran
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ravi Rajaram
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David C Rice
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Garrett L Walsh
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
4
|
Boussios S, Sheriff M, Rassy E, Moschetta M, Samartzis EP, Hallit R, Sadauskaite A, Katsanos KH, Christodoulou DK, Pavlidis N. Immuno-oncology: a narrative review of gastrointestinal and hepatic toxicities. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:423. [PMID: 33842644 PMCID: PMC8033350 DOI: 10.21037/atm-20-7361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccines, cytokines, and adoptive cellular therapies (ACT) represent immuno-therapeutic modalities with great development potential, and they are currently approved for the treatment of a limited number of advanced malignancies. The most up-to-date knowledge on the regulation of the anti-cancer immune response has recently led to the development and approval of inhibitors of immune checkpoints, which have produced unprecedented clinical activity in several hard to treat solid malignancies. However, severe adverse events (AEs) represent a limitation to the use of these drugs. Currently approved checkpoint inhibitors block cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1) and its ligand (PD-L1), resulted in increased survival of patients with several solid and hematologic malignancies. The most common treatment AEs associated with these drugs are fatigue, rash, and auto-immune/inflammatory reactions. Many of the immune-related AEs are reversible and the strategies for their management include supportive care either with or without treatment withdrawal; nevertheless, in severe cases, hospitalization and treatment with immune suppressants, and/or immunomodulators may be required. Steroid therapy is a critical component of the treatment algorithm; nevertheless, the associated immunosuppression may compromise the antitumor response. This article provides a comprehensive and narrative review of luminal gastrointestinal and hepatic complications, including recommendations for their investigation and management.
Collapse
Affiliation(s)
- Stergios Boussios
- King's College London, Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, SE1 9RT, London, UK.,Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, UK.,AELIA Organization, 9th Km Thessaloniki - Thermi, Thessaloniki, Greece
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent, ME7 5NY, UK
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy Institut, Villejuif, France.,Department of Hematology-Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Michele Moschetta
- CHUV, Lausanne University Hospital, Rue du Bugnon 21 CH-1011, Lausanne, Switzerland
| | - Eleftherios P Samartzis
- Department of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zurich, Switzerland
| | - Rachel Hallit
- Gastroenterology Department, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris and University of Paris, Paris, France
| | - Agne Sadauskaite
- Department of Pharmacy, Medway NHS Foundation Trust, Gillingham, Kent, UK
| | - Konstantinos H Katsanos
- Department of Gastroenterology, University Hospital of Ioannina, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dimitrios K Christodoulou
- Department of Gastroenterology, University Hospital of Ioannina, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | | |
Collapse
|
5
|
Mohr P, Kiecker F, Soriano V, Dereure O, Mujika K, Saiag P, Utikal J, Koneru R, Robert C, Cuadros F, Chacón M, Villarroel RU, Najjar YG, Kottschade L, Couselo EM, Koruth R, Guérin A, Burne R, Ionescu-Ittu R, Perrinjaquet M, Zager JS. Adjuvant therapy versus watch-and-wait post surgery for stage III melanoma: a multicountry retrospective chart review. Melanoma Manag 2019; 6:MMT33. [PMID: 31871622 PMCID: PMC6923782 DOI: 10.2217/mmt-2019-0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022] Open
Abstract
AIM To describe treatment patterns among patients with stage III melanoma who underwent surgical excision in years 2011-2016, and assess outcomes among patients who subsequently received systemic adjuvant therapy versus watch-and-wait. METHODS Chart review of 380 patients from 17 melanoma centers in North America, South America and Europe. RESULTS Of 129 (34%) patients treated with adjuvant therapy, 85% received interferon α-2b and 56% discontinued treatment (mostly due to adverse events). Relapse-free survival was significantly longer for patients treated with adjuvant therapy versus watch-and-wait (hazard ratio = 0.63; p < 0.05). There was considerable heterogeneity in adjuvant treatment schedules and doses. Similar results were found in patients who received interferon-based adjuvant therapy. CONCLUSION Adjuvant therapies with better safety/efficacy profiles will improve clinical outcomes in patients with stage III melanoma.
Collapse
Affiliation(s)
- Peter Mohr
- Department of Dermatology, Elbe Kliniken, Stade, Germany
| | - Felix Kiecker
- Department of Dermatology and Allergy, Skin Cancer Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Virtudes Soriano
- Department of Medical Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Olivier Dereure
- Department of Dermatology and INSERM U1058 ‘pathogenesis and control of chronic infections’, University of Montpellier, Montpellier, France
| | - Karmele Mujika
- Department of Medical Oncology, Onkologikoa-Oncology Institute Gipuzkoa, Gipuzkoa, Spain
| | - Philippe Saiag
- Department of General and Oncologic Dermatology Ambroise Paré Hospital, APHP; EA 4340 ‘Biomarkers in cancerology and hemato-oncology’, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany and Department of Dermatology, Venereology and Allergology; University Medical Center, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Rama Koneru
- RS McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, Oshawa, Ontario, Canada
| | - Caroline Robert
- Dermatology Unit, Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - Florencia Cuadros
- Medical Oncology, Instituto de Oncologia de Rosario, Rosario, Santa Fe, Argentina
| | - Matias Chacón
- Departments of Medical and Surgical Oncology, Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | - Yana G Najjar
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lisa Kottschade
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Eva M Couselo
- Department of Medical Oncology, Vall d'Hebron Hospital and VHIO (Vall d'Hebron Institute of Oncology), Barcelona, Spain
| | - Roy Koruth
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | | | | | | | | | - Jonathan S Zager
- Departments of Cutaneous Oncology and Sarcoma, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
6
|
Abstract
Interferon gamma has long been studied as a critical mediator of tumor immunity. In recent years, the complexity of cellular interactions that take place in the tumor microenvironment has become better appreciated in the context of immunotherapy. While checkpoint inhibitors have dramatically improved remission rates in cancer treatment, IFN-γ and related effectors continue to be identified as strong predictors of treatment success. In this review, we provide an overview of the multiple immunosuppressive barriers that IFN-γ has to overcome to eliminate tumors, and potential avenues for modulating the immune response in favor of tumor rejection.
Collapse
Affiliation(s)
- J Daniel Burke
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
7
|
Lau E, Sedy J, Sander C, Shaw MA, Feng Y, Scortegagna M, Claps G, Robinson S, Cheng P, Srivas R, Soonthornvacharin S, Ideker T, Bosenberg M, Gonzalez R, Robinson W, Chanda SK, Ware C, Dummer R, Hoon D, Kirkwood JM, Ronai ZA. Transcriptional repression of IFNβ1 by ATF2 confers melanoma resistance to therapy. Oncogene 2015; 34:5739-48. [PMID: 25728676 PMCID: PMC4558399 DOI: 10.1038/onc.2015.22] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/02/2015] [Accepted: 01/06/2015] [Indexed: 02/07/2023]
Abstract
The resistance of melanoma to current treatment modalities represents a major obstacle for durable therapeutic response, and thus, the elucidation of mechanisms of resistance is urgently needed. The crucial functions of Activating Transcription Factor-2 (ATF2) in the development and therapeutic resistance of melanoma have been previously reported, although the precise underlying mechanisms remain unclear. Here, we report a protein kinase C epsilon (PKCε)- and Activating Transcription Factor-2 (ATF2)-mediated mechanism that facilitates resistance by transcriptionally repressing the expression of IFNβ1 and downstream type-I IFN signaling, which is otherwise induced upon exposure to chemotherapy. Treatment of early stage melanomas expressing low levels of PKCε with chemotherapies relieves its transcriptional repression of IFNB1, resulting in impaired S-phase progression, a senescence-like phenotype, and increased cell death. This response is lost in late stage metastatic melanomas expressing high levels of PKCε. Notably, nuclear ATF2 and low expression of IFNβ1 in melanoma tumor samples correlates with poor patient responsiveness to biochemotherapy or neoadjuvant IFN-α2a. Conversely, cytosolic ATF2 and induction of IFNβ1 coincides with therapeutic responsiveness. Collectively, we identify an IFNβ1-dependent, cell autonomous mechanism that contributes to the therapeutic resistance of melanoma via the PKCε-ATF2 regulatory axis.
Collapse
Affiliation(s)
- E Lau
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - J Sedy
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - C Sander
- University of Pittsburgh Cancer Center, Pittsburgh, PA, USA
| | - M A Shaw
- John Wayne Cancer Institute, Santa Monica, CA, USA
| | - Y Feng
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - M Scortegagna
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - G Claps
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - S Robinson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - P Cheng
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - R Srivas
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - S Soonthornvacharin
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - T Ideker
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | | | - R Gonzalez
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - W Robinson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - S K Chanda
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - C Ware
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - R Dummer
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - D Hoon
- John Wayne Cancer Institute, Santa Monica, CA, USA
| | - J M Kirkwood
- University of Pittsburgh Cancer Center, Pittsburgh, PA, USA
| | - Z A Ronai
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
8
|
|
9
|
Alatrash G, Jakher H, Stafford PD, Mittendorf EA. Cancer immunotherapies, their safety and toxicity. Expert Opin Drug Saf 2013; 12:631-45. [DOI: 10.1517/14740338.2013.795944] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
10
|
Curigliano G, Locatelli M, Fumagalli L, Brollo J, Munzone E, Nolé F, Criscitiello C, Goldhirsch A. Targeting the subtypes of breast cancer: rethinking investigational drugs. Expert Opin Investig Drugs 2012; 21:191-204. [DOI: 10.1517/13543784.2012.651456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
Fernandez AA, Paniker L, Garcia R, Mitchell DL. Recent advances in sunlight-induced carcinogenesis using the Xiphophorus melanoma model. Comp Biochem Physiol C Toxicol Pharmacol 2012; 155:64-70. [PMID: 21457786 PMCID: PMC3164944 DOI: 10.1016/j.cbpc.2011.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 03/23/2011] [Accepted: 03/23/2011] [Indexed: 01/25/2023]
Abstract
Unlike breast and prostate cancers, the nature and sequence of critical genetic and epigenetic events involved in the initiation and progression of melanoma are not well understood. A contributing factor to this dilemma, especially given our current understanding of the importance of UV light in melanoma etiology, is the lack of quality UV-inducible melanoma animal models. In this study we elaborate on the capability of UV light to induce cutaneous malignant melanomas (CMM) in Xiphophorus fishes, which were previously found to develop melanomas after acute neonatal UVB irradiation. In two separate tumorigenesis experiments, we exposed adult Xiphophorus hybrids to either acute UVB irradiations (5 consecutive daily treatments) or chronic solar irradiations (continuous UVA/UVB treatment for 9 months). Acute adult UVB irradiation resulted in the significant induction of melanomas, and moreover, this induction rate is equivalent to that of animals exposed to acute neonatal UVB irradiation. This study represents the first evidence that acute adult UVB irradiation, in the absence of any early life exposures, induces CMM. Similar to the findings conducted on other divergent melanoma models, including HGF/SF transgenic mice and Monodelphis domestica, prolonged chronic solar UV was not a factor in melanomagenesis.
Collapse
Affiliation(s)
- André A Fernandez
- The University of Texas MD Anderson Cancer Center, Department of Carcinogenesis, 1808 Park Road 1C, Smithville, TX 78957, USA.
| | | | | | | |
Collapse
|
12
|
Hauschild A. Adjuvant interferon alfa for melanoma: new evidence-based treatment recommendations? ACTA ACUST UNITED AC 2011; 16:3-6. [PMID: 19526078 PMCID: PMC2695706 DOI: 10.3747/co.v16i3.447] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- A Hauschild
- Axel Hauschild, Department of Dermatology, University of Kiel, Schittenhelmstr. 7, Kiel, 24105, Germany.
| |
Collapse
|
13
|
Kaur B, Lesinski GB, Chaudhury AR. From Concept to the Clinics: Development of Novel Large Molecule Cancer Therapeutics. PHARMACEUTICAL SCIENCES ENCYCLOPEDIA 2010. [DOI: 10.1002/9780470571224.pse402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
14
|
Kashani-Sabet M, Venna S, Nosrati M, Rangel J, Sucker A, Egberts F, Baehner FL, Simko J, Leong SPL, Haqq C, Hauschild A, Schadendorf D, Miller JR, Sagebiel RW. A multimarker prognostic assay for primary cutaneous melanoma. Clin Cancer Res 2009; 15:6987-92. [PMID: 19887476 DOI: 10.1158/1078-0432.ccr-09-1777] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE To determine the prognostic significance of a multimarker assay incorporating expression levels of three molecular markers in primary cutaneous melanoma. EXPERIMENTAL DESIGN We assessed expression levels of NCOA3, SPP1, and RGS1 using immunohistochemical analysis in a tissue microarray cohort of 395 patients. For each marker, we identified optimal cut-points for expression intensity to predict disease-specific survival (DSS) and, as a secondary endpoint, sentinel lymph node (SLN) status. The cumulative overexpression of all three markers was embodied in a multimarker index, and its prognostic effect on DSS and SLN status was assessed using Cox regression, Kaplan-Meier analysis, and logistic regression. The prognostic effect of this multimarker assay on DSS was assessed in an independent cohort of 141 patients, in which marker expression levels were scored using immunohistochemical analysis of stained tissue sections. RESULTS Increasing multimarker index scores were significantly predictive of reduced DSS and increased SLN metastasis in the 395-patient cohort. Multivariate logistic regression analysis revealed multimarker expression scores as an independent predictor of SLN status (P = 0.001). Multivariate Cox regression analysis showed the independent effect of the multimarker index on DSS (P < 0.001). The multimarker index was the most significant factor predicting DSS when compared with other clinical and histologic factors, including SLN status (P = 0.002). Multimarker expression scores were also the most significantly predictive of DSS in the independent cohort (P = 0.01). CONCLUSIONS These results describe a multimarker assay with independent prognostic effect on the prediction of survival associated with melanoma in two distinct cohorts.
Collapse
Affiliation(s)
- Mohammed Kashani-Sabet
- Auerback Melanoma Research Laboratory and Melanoma Center, Comprehensive Cancer Center, and Departments of Pathology, Surgery, and Urology, University of California-San Francisco, San Francisco, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Self-renewing cancer stem cells (CSC) capable of spawning more differentiated tumor cell progeny are required for tumorigenesis and neoplastic progression of leukemias and several solid cancers. The mechanisms by which CSC cause tumor initiation and growth are currently unknown. Recent findings that suggest a negative correlation between degrees of host immunocompetence and rates of cancer development raise the possibility that only a restricted minority of malignant cells, namely CSC, may possess the phenotypic and functional characteristics to evade host antitumor immunity. In human malignant melanoma, a highly immunogenic cancer, we recently identified malignant melanoma initiating cells (MMIC), a novel type of CSC, based on selective expression of the chemoresistance mediator ABCB5. Here we present evidence of a relative immune privilege of ABCB5(+) MMIC, suggesting refractoriness to current immunotherapeutic treatment strategies. We discuss our findings in the context of established immunomodulatory functions of physiologic stem cells and in relation to mechanisms responsible for the downregulation of immune responses against tumors. We propose that the MMIC subset might be responsible for melanoma immune evasion and that immunomodulation might represent one mechanism by which CSC advance tumorigenic growth and resistance to immunotherapy. Accordingly, the possibility of an MMIC-driven tumor escape from immune-mediated rejection has important implications for current melanoma immunotherapy.
Collapse
Affiliation(s)
- Tobias Schatton
- Transplantation Research Center, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
16
|
Eggermont AMM, Bouwhuis MG, Kruit WH, Testori A, ten Hagen T, Yver A, Xu C. Serum concentrations of pegylated interferon α-2b in patients with resected stage III melanoma receiving adjuvant pegylated interferon α-2b in a randomized phase III trial (EORTC 18991). Cancer Chemother Pharmacol 2009; 65:671-7. [DOI: 10.1007/s00280-009-1072-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 07/07/2009] [Indexed: 10/20/2022]
|
17
|
Campos SM. Anti-epidermal growth factor receptor strategies for advanced breast cancer. Cancer Invest 2008; 26:757-68. [PMID: 18853311 DOI: 10.1080/07357900801971040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Susana M Campos
- Department of Breast and Gynecology, Dana Farber Cancer Institute, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Di Leo A, Gomez HL, Aziz Z, Zvirbule Z, Bines J, Arbushites MC, Guerrera SF, Koehler M, Oliva C, Stein SH, Williams LS, Dering J, Finn RS, Press MF. Phase III, double-blind, randomized study comparing lapatinib plus paclitaxel with placebo plus paclitaxel as first-line treatment for metastatic breast cancer. J Clin Oncol 2008; 26:5544-52. [PMID: 18955454 DOI: 10.1200/jco.2008.16.2578] [Citation(s) in RCA: 338] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Lapatinib, a dual tyrosine kinase inhibitor of epidermal growth factor receptor (EGFR/ErbB1) and human epidermal growth factor receptor 2 (HER-2/ErbB2), is effective against HER-2-positive locally advanced or metastatic breast cancer (MBC). This phase III trial evaluated the efficacy of lapatinib in HER-2-negative and HER-2-uncharacterized MBC. PATIENTS AND METHODS Women with MBC were randomly assigned to first-line therapy with paclitaxel 175 mg/m(2) every 3 weeks plus lapatinib 1,500 mg/d or placebo. A preplanned retrospective evaluation of HER-2 status was performed using fluorescence in situ hybridization and immunohistochemistry. The primary end point was time to progression (TTP); secondary end points were objective response rate (ORR), clinical benefit rate (CBR), event-free survival (EFS), and overall survival (OS). RESULTS In the intent-to-treat population (n = 579), there were no significant differences in TTP, EFS, or OS between treatment arms, although differences in ORR and CBR were noted. In 86 HER-2-positive patients (15%), treatment with paclitaxel-lapatinib resulted in statistically significant improvements in TTP, EFS, ORR, and CBR compared with paclitaxel-placebo. No differences between treatment groups were observed for any end point in HER-2-negative patients. The most common adverse events were alopecia, rash, and diarrhea. The incidence of diarrhea and rash was significantly higher in the paclitaxel-lapatinib arm. The rate of cardiac events was low, and no difference was observed between treatment arms. CONCLUSION Patients with HER-2-negative or HER-2-untested MBC did not benefit from the addition of lapatinib to paclitaxel. However, first-line therapy with paclitaxel-lapatinib significantly improved clinical outcomes in HER-2-positive patients. Prospective evaluation of the efficacy and safety of this combination is ongoing in early and metastatic HER-2-positive breast cancer patients.
Collapse
Affiliation(s)
- Angelo Di Leo
- Sandro Pitigliani Medical Oncology Unit, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy 59100.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Albain KS, Nag SM, Calderillo-Ruiz G, Jordaan JP, Llombart AC, Pluzanska A, Rolski J, Melemed AS, Reyes-Vidal JM, Sekhon JS, Simms L, O'Shaughnessy J. Gemcitabine plus Paclitaxel versus Paclitaxel monotherapy in patients with metastatic breast cancer and prior anthracycline treatment. J Clin Oncol 2008; 26:3950-7. [PMID: 18711184 DOI: 10.1200/jco.2007.11.9362] [Citation(s) in RCA: 289] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The objective of this phase III global study was to compare the efficacy of gemcitabine plus paclitaxel (GT) versus paclitaxel in patients with advanced breast cancer. It was designed as a pivotal study for the approval of G for a breast cancer treatment indication. PATIENTS AND METHODS Patients who relapsed after adjuvant anthracyclines were randomly assigned to gemcitabine,1,250 mg/m(2) days 1 and 8 plus paclitaxel, 175 mg/m(2) on day 1; or, to paclitaxel at same dose on day 1 (both arms administered every 21 days, unblinded). The primary end point was overall survival (OS) and secondary end points were time to progression (TTP), response rate (RR), progression-free survival, response duration, and toxicity. This final OS analysis was planned at 380 deaths. RESULTS A total of 266 patients were randomly assigned to GT and 263 to paclitaxel. Median survival on GT was 18.6 months versus 15.8 months on paclitaxel (log-rank P = . 0489), with an adjusted Cox hazard ratio of 0.78 (95% CI, 0.64 to 0.96; P = .0187). The TTP was longer (6.14 v 3.98 months; log-rank P = .0002) and the RR was better (41.4% v 26.2%; P = .0002) on GT. There was more grade 3 to 4 neutropenia on GT and grade 2 to 4 fatigue and neuropathy were slightly more prevalent on GT. CONCLUSION This phase III study documents a role for gemcitabine in advanced breast cancer after anthracycline-based adjuvant therapy. The results establish GT as a reasonable choice for women who require cytoreduction with manageable toxicities and validate ongoing testing of GT in the adjuvant setting.
Collapse
Affiliation(s)
- Kathy S Albain
- Loyola University Chicago Stritch School of Medicine, Cardinal Bernardin Cancer Center, 2160 S First Ave, Maywood, IL 60153, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Affiliation(s)
- Vernon K Sondak
- H Lee Moffitt Cancer Center and University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | - Lawrence E Flaherty
- Karmanos Cancer Institute and Wayne State University School of Medicine, MI, Detroit, USA
| |
Collapse
|
21
|
Zimmerer JM, Lehman AM, Ruppert AS, Noble CW, Olencki T, Walker MJ, Kendra K, Carson WE. IFN-α-2b–Induced Signal Transduction and Gene Regulation in Patient Peripheral Blood Mononuclear Cells Is Not Enhanced by a Dose Increase from 5 to 10 Megaunits/m2. Clin Cancer Res 2008; 14:1438-45. [DOI: 10.1158/1078-0432.ccr-07-4178] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
El-Maraghi RH, Eisenhauer EA. Review of phase II trial designs used in studies of molecular targeted agents: outcomes and predictors of success in phase III. J Clin Oncol 2008; 26:1346-54. [PMID: 18285606 DOI: 10.1200/jco.2007.13.5913] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Because the appropriate design and end points for phase II evaluation of targeted anticancer agents are unclear, we undertook a review of recent reports of phase II trials of targeted agents to determine the types of designs used, the planned end points, the outcomes, and the relationship between trial outcomes and regulatory approval. METHODS We retrieved reports of single-agent phase II trials in six solid tumors for 19 targeted drugs. For each, we abstracted data regarding planned design and actual results. Response rates were examined for any relationship to eventual success of the agents, as determined by US Food and Drug Administration approval for at least one indication. RESULTS Eighty-nine trials were identified. Objective response was the primary or coprimary end point in the majority of trials (61 of 89 trials). Fourteen reports were of randomized studies generally evaluating different doses of agents, not as controlled experiments. Enrichment for target expression was uncommon. Objective responses were seen in 38 trials; in 19 trials, response rates were more than 10%, and in eight, they were more than 20%. Agents with high response rates tended to have high nonprogression rates; renal cell carcinoma was the exception to this. Higher overall response rates were predictive of regulatory approval in the tumor types reviewed (P = .005). CONCLUSION In practice, phase II design for targeted agents is similar to that for cytotoxics. Objective response seems to be a useful end point for screening new targeted agents because, in our review, its observation predicted for eventual success. Improvements in design are recommended, as is more frequent inclusion of biological questions as part of phase II trials.
Collapse
|
23
|
Melanoma and Other Cutaneous Malignancies. Surgery 2008. [DOI: 10.1007/978-0-387-68113-9_98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
24
|
Cutaneous Melanoma. Oncology 2007. [DOI: 10.1007/0-387-31056-8_59] [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]
|
25
|
Nieto Y, Nawaz F, Jones RB, Shpall EJ, Nawaz S. Prognostic significance of overexpression and phosphorylation of epidermal growth factor receptor (EGFR) and the presence of truncated EGFRvIII in locoregionally advanced breast cancer. J Clin Oncol 2007; 25:4405-13. [PMID: 17906204 DOI: 10.1200/jco.2006.09.8822] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The prognostic value of the epidermal growth factor receptor (EGFR) in breast cancer and more specifically, in patients with locoregionally advanced disease, is still undefined. We hypothesized that EGFR status plays a major prognostic role in this setting, through expression, activation, or the presence of its mutated variant EGFRvIII. PATIENTS AND METHODS We reviewed tumor samples of 225 patients treated uniformly in prospective trials of high-dose chemotherapy for four to nine positive axillary nodes, > or = 10 positive nodes, or inflammatory carcinoma, and observed for a median of 9 years (range, 3 to 13 years). We analyzed the effect on outcome of expression of EGFR, phosphorylated EGFR (phospho-EGFR), and EGFRvIII, as studied by immunohistochemistry. RESULTS EGFR expression, phospho-EGFR, and mutated EGFRvIII were detected in 43%, 54%, and 4% of the patients, respectively. EGFR expression correlated with negative hormone receptor status, and was associated with significantly worse relapse-free survival (59% v 79%; P < .001) and overall survival (61% v 81%; P = .001) than no expression. There was no association of phospho-EGFR or EGFRvIII with outcome. Multivariate models confirmed the prognostic effect of EGFR independent of other known prognostic variables in this population. The prognostic value of EGFR was most prominent in the human epidermal growth factor receptor 2 (HER-2) -positive and the estrogen receptor/progesterone receptor-negative subgroups. CONCLUSION EGFR expression, but not phospho-EGFR or EGFRvIII expression, is an independent adverse prognostic factor in patients with high-risk primary breast cancer, particularly when it is coexpressed with HER-2. Our results suggest the potential benefit of dual EGFR/HER-2 receptor targeting in this setting.
Collapse
Affiliation(s)
- Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, Unit 423, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
26
|
Cormier JN, Xing Y, Ding M, Cantor SB, Salter KJ, Lee JE, Mansfield PF, Gershenwald JE, Ross MI. Cost Effectiveness of Adjuvant Interferon in Node-Positive Melanoma. J Clin Oncol 2007; 25:2442-8. [PMID: 17557957 DOI: 10.1200/jco.2007.10.7284] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To assess the benefits of adjuvant high-dose interferon alfa (HDI) treatment for patients with high-risk melanoma. Methods We designed a decision-analytic probabilistic Markov model to simulate the natural history of patients with stage IIIA, IIIB, and IIIC melanoma and evaluate the outcomes with and without HDI treatment. Outcomes were in quality-adjusted life years (QALYs) and incremental cost-effectiveness ratios (ICERs). Probability estimates and costs were derived from primary patient level data, while the efficacy of HDI and associated utilities were determined from published reports. The base-case analysis was a cohort of 50-year-old patients. Results HDI increased the median life expectancy in patients with stage III melanoma from 3.75 years in the observation cohort to 4.42 years in the HDI cohort. The difference in QALYs ranged from 0.31 years for stage IIIA patients to 0.60 years for stage IIIC patients treated with HDI. HDI was cost effective in patients with stage IIIB and IIIC melanoma, with ICERs of $95,304 and $76,068 per QALY gained, respectively. Using a threshold of $100,000 per QALY gained, HDI was cost effective for all stage III patients younger than 52 years. HDI was not cost effective for patients with stage IIIA disease and for subsets of patients older than 63 years with stage IIIB disease. Conclusion Our model demonstrates that the probability of HDI being cost effective varies substantially by melanoma substage. HDI showed the greatest benefit in terms of QALYs and was most cost effective in patients younger than 60 years with stage IIIC disease.
Collapse
Affiliation(s)
- Janice N Cormier
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77230-1402, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW This review summarizes the continuing value of some therapeutic drugs and new agents under development for the treatment of breast cancer. RECENT FINDINGS Overexpression and activation of various growth factor receptors occurs frequently in human breast cancer. Therapeutic approaches mainly involve the epidermal growth factor receptor family, insulin-like growth factor receptor and vascular endothelial growth factor receptor. Therapeutic agents targeting these receptors include the monoclonal antibodies trastuzumab and pertuzumab, and the small-molecule inhibitors gefitinib and erlotinib. Other small-molecule and dual inhibitors are in development, some of which have been demonstrated to have higher efficacy in the treatment of breast cancer. The selective estrogen receptor modulators and aromatase inhibitors continue to be valuable in the endocrine therapy of breast cancer. These drugs have been shown to have higher efficacy than conventional therapy agents, and to have extensive potential, especially in the treatment of postmenopausal women with advanced breast cancer. SUMMARY Approved agents including epidermal growth factor receptor-targeted inhibitor, selective estrogen receptor modulators and aromatase inhibitors continue to be valuable in treating breast cancer. To overcome the acquired resistance caused by these agents and to enhance the therapy effect, the development of new and specific dual inhibitors targeting various growth factor receptors will be important in the future.
Collapse
Affiliation(s)
- Xiao-ping Gao
- Institute of Materia Medica, KangHong Pharmaceutical Goup, Chengdu, People's Republic of China.
| | | |
Collapse
|
28
|
On the appropriateness of appropriateness judgments: The case of interferon treatment for melanoma. JUDGMENT AND DECISION MAKING 2007. [DOI: 10.1017/s1930297500000292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AbstractWe compare experts’ judgments of the appropriateness of a treatment (interferon treatment for melanoma) on the basis of important attributes of this disease (thickness, ulceration, lymph node involvement and type of metastases) to a decision analytic model in which the probabilities of deterioration are derived from the medical literature and from epidemiological studies. The comparison is based on what we call the linearity test, which examines whether appropriateness judgments are a linear function of the epidemiological value of p2, the probability of deterioration of the patient condition if he would have received the treatment. This comparison allows for the assessment of the validity of the experts’ judgments under the assumption that the decision analytic model is valid, or alternatively, the assessment of the validity of the decision analytic model under the assumption that the experts’ judgments are valid. Under the former assumption the results indicate that appropriateness judgments are by and large accurate. Under the latter assumption the results support the idea of a constant treatment effect, the idea that efficacy of a treatment is constant over various levels of severity of the disease. Our results also support the idea that experts’ aggregate judgments far exceed individuals’ judgments.
Collapse
|
29
|
Quesada AR, Muñoz-Chápuli R, Medina MA. Anti-angiogenic drugs: from bench to clinical trials. Med Res Rev 2006; 26:483-530. [PMID: 16652370 DOI: 10.1002/med.20059] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Angiogenesis, the generation of new capillaries through a process of pre-existing microvessel sprouting, is under stringent control and normally occurs only during embryonic and post-embryonic development, reproductive cycle, and wound repair. However, in many pathological conditions (solid tumor progression, metastasis, diabetic retinopathy, hemangioma, arthritis, psoriasis and atherosclerosis among others), the disease appears to be associated with persistent upregulated angiogenesis. The development of specific anti-angiogenic agents arises as an attractive therapeutic approach for the treatment of cancer and other angiogenesis-dependent diseases. The formation of new blood vessels is a complex multi-step process. Endothelial cells resting in the parent vessels are activated by an angiogenic signal and stimulated to synthesize and release degradative enzymes allowing endothelial cells to migrate, proliferate and finally differentiate to give rise to capillary tubules. Any of these steps may be a potential target for pharmacological intervention. In spite of the disappointing results obtained initially in clinical trials with anti-angiogenic drugs, recent reports with positive results in phases II and III trials encourage expectations in their therapeutic potential. This review discusses the current approaches for the discovery of new compounds that inhibit angiogenesis, with emphasis on the clinical developmental status of anti-angiogenic drugs.
Collapse
Affiliation(s)
- Ana R Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Science, University of Málaga, 29071 Málaga, Spain.
| | | | | |
Collapse
|
30
|
Molenkamp BG, van Leeuwen PAM, van den Eertwegh AJM, Sluijter BJR, Scheper RJ, Meijer S, de Gruijl TD. Immunomodulation of the melanoma sentinel lymph node: a novel adjuvant therapeutic option. Immunobiology 2006; 211:651-61. [PMID: 16920504 DOI: 10.1016/j.imbio.2006.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cutaneous melanoma is the most aggressive type of skin cancer. Paradoxically, melanoma is also the most immunogenic tumour identified to date: tumour-reactive T cells are detectable both in the blood and in tumour-draining lymph nodes (TDLN) of melanoma patients and their frequency can be increased by specific vaccination. However, early melanoma development is accompanied by impaired immune effector functions in the initial TDLN, the sentinel lymph node (SLN). Most notably, a reduced frequency and activation state of dendritic cells (DC) interferes with the uptake and presentation of tumour-associated antigens (TAA) to specific anti-tumour cytotoxic T-lymphocytes (CTL) and T helper cells (Th). These impaired immune effector functions may contribute to the early metastatic events that are associated with this tumour type. Since complete surgical excision at an early stage remains the only curative treatment option (adjuvant therapy options are limited and show no survival benefits), immunopotentiation of the SLN to jump-start or boost tumour specific immunity in early stage melanoma may be a valuable adjuvant treatment option that can be generally applied with minimal discomfort to the patient. Early clinical studies indicate that local Granulocyte/Macrophage-Colony Stimulating Factor (GM-CSF) or Cytosine-phosphate-Guanine (CpG) administration leads to activation of different DC subsets and conditions the SLN microenvironment to be more conducive to the generation of T-cell-mediated anti-tumour immunity.
Collapse
Affiliation(s)
- Barbara G Molenkamp
- Department of Surgical Oncology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Lens M. Cutaneous melanoma: interferon alpha adjuvant therapy for patients at high risk for recurrent disease. Dermatol Ther 2006; 19:9-18. [PMID: 16405565 DOI: 10.1111/j.1529-8019.2005.00051.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemic adjuvant therapy in melanoma patients is the systemic treatment that is administered with the goal of eradicating micrometastatic deposits in patients who are clinically free of disease after surgical removal of the primary melanoma, but with a high risk of systemic recurrence. Interferon-alpha (IFN-alpha) is one of the most frequently used adjuvant therapies. Several randomized trials evaluated the efficacy of IFN-alpha in melanoma patients. However, results from conducted trials are controversial. Twelve randomized IFN-alpha trials are discussed in detail. All trials, including meta-analysis, failed to demonstrate a clear impact of IFN-alpha therapy on overall survival in melanoma patients. Based on currently available evidence, IFN-alpha therapy in the adjuvant setting should not be considered standard of care for patients who have melanoma. Results from ongoing studies are awaited. Further research for this therapy is required.
Collapse
Affiliation(s)
- Marko Lens
- Imperial College, Department of Epidemiology & Public Health, Faculty of Medicine, St Mary's Campus, Norfolk Place, London, England.
| |
Collapse
|
32
|
Richtig E, Langmann G, Schlemmer G, Müllner K, Papaefthymiou G, Bergthaler P, Smolle J. Verträglichkeit und Wirksamkeit einer adjuvanten Interferon-alfa-2b-Behandlung beim Aderhautmelanom. Ophthalmologe 2006; 103:506-11. [PMID: 16763868 DOI: 10.1007/s00347-006-1350-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE In this manuscript, the safety and efficacy of adjuvant interferon alfa 2b treatment of uveal melanoma is described. PATIENTS AND METHODS A total of 39 patients (23 male and 16 female, mean age 56.5 years, range 35-78 years) with uveal melanoma were treated with interferon alfa 2b, 3 million units three times a week subcutaneously for 1 year after therapy of the primary tumor. In all patients age, gender, primary melanoma data, therapeutic interventions, treatment side effects and outcome were documented. RESULTS Of the 39 patients, 31 (80%) finished the treatment as scheduled after 1 year. In 18 patients (46%) the initial dose had to be reduced due to leucopenia, thrombopenia, cardiac symptoms, elevated of liver function or vertigo (WHO grade I-III). In eight patients, therapy had to be withdrawn because of serious side effects (five patients) and the appearance of metastases (three patients). Neither a univariate approach nor a multivariate approach could show a protective effect of interferon treatment on survival. CONCLUSIONS Adjuvant treatment of uveal melanoma with interferon alfa should be abandoned until the question of dose and administration for cutaneous melanoma is solved.
Collapse
Affiliation(s)
- E Richtig
- Abt. für Allgemeine Dermatologie, Universitätsklinik für Dermatologie und Venerologie, Medizinische Universität Graz, Auenbruggerplatz 8, 8036 Graz, Austria.
| | | | | | | | | | | | | |
Collapse
|
33
|
Schneider B, Fukunaga A, Murry D, Yoder C, Fife K, Foster A, Rosenberg L, Kelich S, Li L, Sweeney C. A phase I, pharmacokinetic and pharmacodynamic dose escalation trial of weekly paclitaxel with interferon-α2b in patients with solid tumors. Cancer Chemother Pharmacol 2006; 59:261-8. [PMID: 16733646 DOI: 10.1007/s00280-006-0264-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2005] [Accepted: 03/28/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Paclitaxel and interferon have demonstrated anti-angiogenic activity in vitro and in vivo. The toxicity, pharmacokinetics, and pharmacodynamics of paclitaxel with interferon-alpha2b (IFN-alpha2b) were assessed in patients with solid tumors to assess the feasibility of this novel anti-angiogenic regimen. METHODS IFN-alpha2b (1 million units) was administered twice daily by subcutaneous injection. Paclitaxel was given weekly over 1 h starting at 30 mg/m2 and increased to 50 mg/m2. Cycles were repeated every 4 weeks. RESULTS Nineteen patients with a variety of solid tumors were enrolled. Dose-limiting toxicity in cycle 1 was observed at 50 mg/m2. Eleven patients were treated at 40 mg/m2 with no undue toxicity. Pharmacokinetic parameter comparison studies were completed in 11 patients who received days 1 and 29 paclitaxel. Mean paclitaxel clearance and area under the curve (0-infinity) were not statistically different from days 1 to 29. There was a 50% increase in the average Cmax from days 1 to 29. There was also a 73% decrease of matrix metalloproteinase-9 (MMP-9) levels in these 11 patients from days 1 to 29 (p < 0.0005). All three patients with cutaneous angiosarcomas experienced clinically meaningful remissions. In addition, minor responses were observed in one patient with heavily pretreated ovarian cancer and another with adrenocortical carcinoma. CONCLUSION This trial details the inability to dose escalate to the maximum tolerated dose of weekly paclitaxel when combined with low-dose interferon. However, this low-dose regimen caused a significant decrease in MMP-9 and demonstrated anti-cancer activity in cutaneous angiosarcomas.
Collapse
Affiliation(s)
- Bryan Schneider
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
In Reply. Oncologist 2006. [DOI: 10.1634/theoncologist.11-5-539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
35
|
Dummer R, Garbe C, Thompson JA, Eggermont AM, Yoo K, Maier T, Bergstrom B. Randomized dose-escalation study evaluating peginterferon alfa-2a in patients with metastatic malignant melanoma. J Clin Oncol 2006; 24:1188-94. [PMID: 16505439 DOI: 10.1200/jco.2005.04.3216] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE A pegylated interferon, peginterferon alfa-2a (PEG-IFNalpha-2a; 40 kd), has the potential for improved tumor response and survival with lower toxicity than IFNalpha. This open-label, randomized study evaluated the safety, tolerability, and efficacy of subcutaneous PEG-IFNalpha-2a in patients with metastatic malignant melanoma (stage IV American Joint Committee on Cancer staging system). PATIENTS AND METHODS PEG-IFNalpha-2a was administered subcutaneously at 180 (n = 48), 360 (n = 53), or 450 mug (n = 49) once weekly for 24 weeks, with maintenance therapy for responders. Efficacy was assessed by the proportion of patients with complete response (CR) or partial response (PR). RESULTS The major response rate (CR or PR) was 6% in the 180-mug group (CR, 2%; PR, 4%), 8% in the 360-mug group (CR, 2%; PR, 6%), and 12% in the 450-mug group (CR, 6%; PR, 6%). The times to achieve a major response, duration of major response, rate of disease progression, and 12-month survival were similar between groups, although overall median survival was significantly different among the three groups (P = .0136). More patients required dose adjustment for safety reasons in the higher dose groups, but PEG-IFNalpha-2a was generally well tolerated, with few withdrawals because of adverse events (6%, 19%, and 16% in the 180-, 360-, and 450-mug groups, respectively). The most common adverse events were fatigue, pyrexia, and nausea. CONCLUSION PEG-IFNalpha-2a at doses up to 450 mug once weekly has shown good tolerability and similar efficacy to conventional IFNalpha and monochemotherapy in stage IV metastatic melanoma.
Collapse
Affiliation(s)
- Reinhard Dummer
- Department of Dermatology, University Hospital of Zürich, Zürich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
36
|
Chiarion-Sileni V, Del Bianco P, Romanini A, Guida M, Paccagnella A, Dalla Palma M, Naglieri E, Ridolfi R, Silvestri B, Michiara M, De Salvo GL. Tolerability of intensified intravenous interferon alfa-2b versus the ECOG 1684 schedule as adjuvant therapy for stage III melanoma: a randomized phase III Italian Melanoma Inter-group trial (IMI - Mel.A.) [ISRCTN75125874]. BMC Cancer 2006; 6:44. [PMID: 16504154 PMCID: PMC1421423 DOI: 10.1186/1471-2407-6-44] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 02/27/2006] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND High-dose interferon alfa-2b (IFNalfa-2b), according to the ECOG 1684 schedule, is the only approved adjuvant treatment for stage III melanoma patients by the FDA and EMEA. However, the risk/benefit profile has been questioned limiting its world-wide use. In the late nineties, the Italian Melanoma Inter-group started a spontaneous randomized clinical trial (RCT) to verify if a more intense, but shorter than the ECOG 1684 regimen, could improve survival without increasing the toxicity profile. The safety analysis in the first 169 patients who completed the treatment is here described. METHODS Stage III melanoma patients were randomized to receive IFNalfa-2b 20 MU/m2/d intravenously (IV) 5 days/week x 4 weeks, repeated for three times on weeks 9 to 12, 17 to 20, 25 to 28 (Dose-Dense/Dose-Intense, DD/DI, arm), or IFNalfa-2b 20 MU/m2/d IV 5 days/week x 4 weeks followed by 10 MU/m2 subcutaneously (SC) three times per week x 48 weeks (High Dose Interferon, HDI, arm). Toxicity was recorded and graded, according to the WHO criteria, as the worst grade that occurred during each cycle. RESULTS The most common toxicities in both arms were flu-like and gastrointestinal symptoms, leukopenia, liver and neuro-psychiatric morbidities; with regard to severe toxicity, only leukopenia was statistically more frequent in DD/DI arm than in HDI arm (24% vs 9%) (p = 0.0074), yet, this did not cause an increase in the infection risk. Discontinuation of treatment, due to toxicity, was observed in 13 and 17% of the patients in the DD/DI and HDI arm, respectively. The median actual dose intensity delivered in the DD/DI arm (36.4 MU/m2/week) was statistically higher than that delivered in the HDI arm (30.7 MU/m2/week) (p = 0.003). CONCLUSION Four cycles of intravenous high-dose IFNalfa-2b can be safely delivered with an increase in the median dose intensity. Efficacy results from this trial are eagerly awaited.
Collapse
Affiliation(s)
| | - Paola Del Bianco
- Clinical Trials and Biostatistics Unit, Istituto Oncologico Veneto, Padua, Italy
| | | | - Michele Guida
- Medical Oncology Unit, Ospedale Oncologico, Bari, Italy
| | | | | | | | | | | | | | - Gian Luca De Salvo
- Clinical Trials and Biostatistics Unit, Istituto Oncologico Veneto, Padua, Italy
| |
Collapse
|
37
|
Abstract
BACKGROUND High-dose interferon (IFN) is the approved agent for adjuvant treatment of melanoma in the United States. This approval is for high-risk, predominantly stage III patients with cutaneous primaries. There are still decisions to be made in the care of these patients. Also, there are questions about whether the IFN data can be extrapolated to patients with other stages of melanoma and whether adjuvant treatment should be offered to these individuals. Clearly there is room for improvement in this area. METHODS The literature on this topic and ongoing national trials in the United States were reviewed. RESULTS The data are insufficient to recommend other agents in the adjuvant treatment of melanoma outside a clinical trial. Extrapolation of the IFN data to patient populations other than those studied is problematic at best. National trials are available for most patient populations. CONCLUSIONS The adjuvant treatment of choice for melanoma patients is participation in a clinical trial.
Collapse
Affiliation(s)
- David H Lawson
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
38
|
Richtig E, Soyer HP, Posch M, Mossbacher U, Bauer P, Teban L, Svolba G, Wolf IH, Fritsch P, Zelger B, Volc-Platzer B, Gebhart W, Mischer P, Steiner A, Pachinger W, Hintner H, Gschnait F, Rappersberger K, Pilarski P, Pehamberger H. Prospective, Randomized, Multicenter, Double-Blind Placebo-Controlled Trial Comparing Adjuvant Interferon Alfa and Isotretinoin With Interferon Alfa Alone in Stage IIA and IIB Melanoma: European Cooperative Adjuvant Melanoma Treatment Study Group. J Clin Oncol 2005; 23:8655-63. [PMID: 16260701 DOI: 10.1200/jco.2004.00.8128] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The combination of interferon alfa (IFNα) and isotretinoin has shown a direct antiproliferative effect on human melanoma cell lines, but it remained unclear whether this combination is more effective than IFNα alone in patients with metastatic melanoma. We evaluated safety and efficacy of IFNα and isotretinoin compared with IFNα alone as adjuvant treatment in patients with primary malignant melanoma stage IIA and IIB. Patients and Methods In a prospective, randomized, double-blind, placebo-controlled trial, 407 melanoma patients in stage IIA (301 patients) and IIB (106 patients) were randomly assigned to either IFNα and isotretinoin (isotretinoin group; 206 patients) or IFNα and placebo (placebo group; 201 patients) after excision of the primary tumor. IFNα was administered three times a week at a dose of 3 million units subcutaneously for 24 months. Isotretinoin at a dose of 20 mg for patients ≤ 73 kg, 30 mg for patients greater than 73 kg, or placebo daily for 24 months. Results A scheduled interim analysis revealed no significant differences in survival rates, with the isotretinoin group and the placebo group showing 5-year disease-free survival rates of 55% (95% CI, 46% to 65%) and 67% (95% CI, 59% to 75%), respectively, and overall 5-year survival rates of 76% (95% CI, 67% to 84%) and 81% (95% CI, 74% to 88%), respectively. The trial was stopped for futility. Conclusion The addition of isotretinoin to an adjuvant treatment of low-dose IFNα in patients with stage IIA and IIB melanoma had no significant effect on disease-free or overall survival and is therefore not recommended.
Collapse
Affiliation(s)
- Erika Richtig
- Department of Dermatology, Medical University of Graz, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ibrahim NK, Samuels B, Page R, Doval D, Patel KM, Rao SC, Nair MK, Bhar P, Desai N, Hortobagyi GN. Multicenter Phase II Trial of ABI-007, an Albumin-Bound Paclitaxel, in Women With Metastatic Breast Cancer. J Clin Oncol 2005; 23:6019-26. [PMID: 16135470 DOI: 10.1200/jco.2005.11.013] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose ABI-007 is a novel nanoparticle, albumin-bound paclitaxel that is free of solvents. This multicenter phase II study was designed to evaluate the efficacy and safety of ABI-007 for the treatment of metastatic breast cancer (MBC). Patients and Methods Sixty-three women with histologically confirmed and measurable MBC received 300 mg/m2 ABI-007 by intravenous infusion over 30 minutes every 3 weeks without premedication. Forty-eight patients received prior chemotherapy; 39 patients received no prior treatment for metastatic disease. Results Overall response rates (complete or partial responses) were 48% (95% CI, 35.3% to 60.0%) for all patients. For patients who received ABI-007 as first-line and greater than first-line therapy for their metastatic disease, the respective response rates were 64% (95% CI, 49.0% to 79.2%) and 21% (95% CI, 7.1% to 42.1%). Median time to disease progression was 26.6 weeks, and median survival was 63.6 weeks. No severe hypersensitivity reactions were reported despite the lack of premedication. Toxicities observed were typical of paclitaxel and included grade 4 neutropenia (24%), grade 3 sensory neuropathy (11%), and grade 4 febrile neutropenia (5%). Patients received a median of six treatment cycles; 16 patients had 25% dose reductions because of toxicities, and two of these patients had subsequent dose reductions. Conclusion ABI-007, the first biologically interactive albumin-bound form of paclitaxel in the nanoparticle state, uses the natural carrier albumin rather than synthetic solvents to deliver paclitaxel and allows for safe administration of high paclitaxel doses without premedication, resulting in significant antitumor activity in patients with MBC, including those receiving the drug as first-line therapy.
Collapse
Affiliation(s)
- Nuhad K Ibrahim
- Department of Breast Medical Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 424, Houston, TX 77030-4009, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Vogel CL, Tan-Chiu E. Trastuzumab Plus Chemotherapy: Convincing Survival Benefit or Not? J Clin Oncol 2005; 23:4247-50. [PMID: 15911863 DOI: 10.1200/jco.2005.12.903] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
41
|
Abstract
Targeted therapeutic agents in breast cancer are representing a larger proportion of new drugs entering clinical testing. Carcinogenesis is a multistep process characterized by genetic alterations that influence key cellular pathways involved in growth and development. Therefore, there are numerous opportunities for pharmacologic targeting. Hormonal therapy is the prototype of a treatment targeting hormone receptors, and this class of drugs still provides the greatest overall impact on outcome. Even though chemotherapy is considered a cytotoxic and nonspecific therapy, it does modulate many key cellular pathways and therefore shares characteristics of biologic drugs. It is clear that targeted therapies are going to play a greater role in improving survival and quality of life in advanced breast cancer, with trastuzumab (Herceptin) serving as a successful model that is a relatively nontoxic agent associated with survival benefits. However, several challenges to the successful identification and application of therapeutic targets remain. These include the dissection of complicated and interacting biologic pathways and the limitations of preclinical models that will allow for a better prioritization of which drugs and combinations will succeed best in the clinic. Better methods for selecting ideal candidates for therapy need to be based on known modes of action. Mechanisms of intrinsic and acquired resistance need further exploration in order to refine drug design. Toxicities that might result from modulation of the targeted pathway must be expected and fully characterized. Some biologic strategies may need to be tested in less refractory cases, or even in early stages, even though this may be more costly and could raise safety concerns. Fortunately progress in all of these areas is expected with the availability of new technologies and a growing infrastructure for preclinical and clinical testing.
Collapse
Affiliation(s)
- Debu Tripathy
- University of Texas Southwestern Medical Center, Dallas, TX 75390-8852, USA.
| |
Collapse
|
42
|
Baselga J, Albanell J, Ruiz A, Lluch A, Gascón P, Guillém V, González S, Sauleda S, Marimón I, Tabernero JM, Koehler MT, Rojo F. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J Clin Oncol 2005; 23:5323-33. [PMID: 15939921 DOI: 10.1200/jco.2005.08.326] [Citation(s) in RCA: 271] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To evaluate the antitumor activity and pharmacodynamic/biologic effect of gefitinib 500 mg/day monotherapy in patients with previously treated, advanced breast cancer. METHODS In this phase II multicenter trial, the primary objective was assessment of the tumor response rate with gefitinib; secondary objectives included analysis of the pharmacodynamic and biologic profiles in healthy and tumor tissue. RESULTS while phosphorylation of mitogen-activated protein kinase was inhibited in both tissues, gefitinib treatment induced p27 and a decrease in Ki67 in skin but not in tumors. Furthermore, gefitinib did not inhibit the activated form of Akt in the tumors. CONCLUSION This study demonstrates a good correlation between the degree of inhibition of EGFR in skin and in breast tumors. The lack of significant clinical activity of gefitinib in our study population is not due to lack of receptor inhibition in these tumors but rather to lack of EGFR dependence in the tested population.
Collapse
Affiliation(s)
- José Baselga
- Medical Oncology Service, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129 08035 Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tabernero J, Rojo F, Marimón I, Voi M, Albanell J, Guix M, Vázquez F, Carulla J, Cooper M, Andreu J, Van Vreckem A, Bellmunt J, Manne V, Manning JA, Garrido C, Felip E, Del Campo JM, García M, Valverde S, Baselga J. Phase I Pharmacokinetic and Pharmacodynamic Study of Weekly 1-Hour and 24-Hour Infusion BMS-214662, a Farnesyltransferase Inhibitor, in Patients With Advanced Solid Tumors. J Clin Oncol 2005; 23:2521-33. [PMID: 15710949 DOI: 10.1200/jco.2005.00.398] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose BMS-214662 is a potent, nonpeptide, small molecule inhibitor of human farnesyltransferase (FT). We have conducted a phase I pharmacokinetic (PK) and pharmacodynamic study of BMS-214662 administered intravenously weekly with 1- and 24-hour infusions. The objectives were to determine the dose-limiting toxicities and the recommended dose (RD), to describe PKs, and to evaluate the relationships between BMS-214662 exposure, FT inhibition, downstream signaling, and induction of apoptosis in tumor samples. Patients and Methods Patients with advanced solid tumors and adequate organ function were eligible. The dose was escalated according to a modified Fibonacci schedule. Results BMS-214662 was escalated from 56 to 278 mg/m2 in 37 patients in the 1-hour schedule, and from 84 to 492 mg/m2 in 31 patients in the 24-hour schedule. Dose-limiting toxicities included gastrointestinal and renal events. The RDs were 209 mg/m2 and 275 mg/m2 in the 1- and 24-hour schedules, respectively. Five patients (three with breast, one with gastric, and one with renal cell cancer) had clinical benefit from treatment. BMS-214662 exhibited linear PKs with area under the concentration-time curves at the RDs of 27 and 32 μM × h in the 1- and 24-hour schedules, respectively. The pattern of FT inhibition in peripheral-blood mononuclear cells at the RDs was different in the two schedules: high (> 80%) but short-lived (≤ 6 hours) in the 1-hour infusion and moderate (> 40%) but long-lived (24 hours) in the 24-hour infusion. BMS-214662 induced apoptosis in tumors but did not inhibit MAPK signaling. Conclusion BMS-214662 can be safely delivered in both the 1-hour and 24-hour infusions at biologically active doses, with the preclinical, PK, and pharmacodynamic profiles favoring the 24-hour schedule.
Collapse
Affiliation(s)
- Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain. [corrected]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The development of molecular targeted anticancer drugs is rapidly changing cancer therapeutics. However, drug resistance to these novel agents remains a real clinical concern. Reports now indicate that resistance to many of these molecular targeted agents--including hormone therapies, trastuzumab, imatinib, and gefitinib--occurs via common resistance mechanisms. These include 1) inadequate target blockade due to sub-optimal drug delivery; 2) altered target expression at the DNA (gene amplification), mRNA or protein level; 3) an altered target such as a mutated kinase domain; 4) modified target regulating proteins (e.g. altered expression of co-activators and/or co-repressors for nuclear steroid hormone receptors); 5) signalling by alternative proteins (functional redundancy) or different signalling pathways. It is envisioned that the molecular evaluation of clinical anticancer drug resistance, which requires the detailed study of pharmacokinetics, pharmacogenetics and pharmacodynamics, will allow the development of rational reversal strategies and improved patient outcome.
Collapse
Affiliation(s)
- L Vidal
- Institute of Cancer Research and Royal Marsden Hospital, Sutton, UK
| | | | | | | |
Collapse
|
45
|
Hancock B, Wheatley K, Ives N, Gore M. Adjuvant Interferon Therapy for Melanoma. J Clin Oncol 2005; 23:2431; author reply 2431-2. [PMID: 15800339 DOI: 10.1200/jco.2005.99.260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
46
|
Affiliation(s)
| | - Stephanie Land
- University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | | | | |
Collapse
|
47
|
Fabian CJ, Kimler BF. Selective Estrogen-Receptor Modulators for Primary Prevention of Breast Cancer. J Clin Oncol 2005; 23:1644-55. [PMID: 15755972 DOI: 10.1200/jco.2005.11.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Carol J Fabian
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| | | |
Collapse
|
48
|
Baselga J, Arteaga CL. Critical update and emerging trends in epidermal growth factor receptor targeting in cancer. J Clin Oncol 2005; 23:2445-59. [PMID: 15753456 DOI: 10.1200/jco.2005.11.890] [Citation(s) in RCA: 539] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase of the ErbB receptor family that is abnormally activated in many epithelial tumors. The aberrant activation of the EGFR leads to enhanced proliferation and other tumor-promoting activities, which provide a strong rationale to target this receptor family. There are two classes of anti-EGFR agents: monoclonal antibodies (MAbs) directed at the extracellular domain of the receptor and small molecule, adenosine triphosphate-competitive inhibitors of the receptor's tyrosine kinase. Anti-EGFR MAbs have shown antitumor activity in advanced colorectal carcinoma, squamous cell carcinomas of the head and neck, non-small-cell lung cancer (NSCLC) and renal cell carcinomas. The tyrosine kinase inhibitors (TKIs) have a partially different activity profile. They are active against NSCLC, and a specific EGFR inhibitor has shown improvement in survival. Recently, mutations and amplifications of the EGFR gene have been identified in NSCLC and predict for enhanced sensitivity to anti-EGFR TKIs. In addition to specific anti-EGFR TKIs, there are broader acting inhibitors such as dual EGFR HER-2 inhibitors and combined anti-pan-ErbB and antivascular endothelial growth factor receptor inhibitors. Current research efforts are directed at selecting the optimal dose and schedule and identifying predictive factors of response and resistance beyond EGFR gene mutations and/or amplifications. Finally, there is a need for improved strategies to integrate anti-EGFR agents with conventional therapies and to explore combinations with other molecular targeted approaches including other antireceptor therapies, receptor-downstream signaling transduction inhibitors, and targeted approaches interfering with other essential drivers of cancer, such as angiogenesis.
Collapse
Affiliation(s)
- José Baselga
- Medical Oncology Service, Vall d'Hebron Research Institute and Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, Barcelona 08035, Spain.
| | | |
Collapse
|
49
|
Zielinski C, Beslija S, Mrsic-Krmpotic Z, Welnicka-Jaskiewicz M, Wiltschke C, Kahan Z, Grgic M, Tzekova V, Inbar M, Cervek J, Chernozemsky I, Szanto J, Spanik S, Wagnerova M, Ghilezan N, Pawlega J, Vrbanec D, Khamtsov D, Soldatenkova V, Brodowicz T. Gemcitabine, Epirubicin, and Paclitaxel Versus Fluorouracil, Epirubicin, and Cyclophosphamide As First-Line Chemotherapy in Metastatic Breast Cancer: A Central European Cooperative Oncology Group International, Multicenter, Prospective, Randomized Phase III Trial. J Clin Oncol 2005; 23:1401-8. [PMID: 15735116 DOI: 10.1200/jco.2005.12.106] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background The objectives of this phase III trial were to compare the time to progressive disease (TtPD), overall response rate (ORR), overall survival, and toxicity of gemcitabine, epirubicin, and paclitaxel (GET) versus fluorouracil (FU), epirubicin, and cyclophosphamide (FEC) as first-line therapy in patients with metastatic breast cancer (MBC). Patients and Methods Female patients aged 18 to 75 years with stage IV and measurable MBC were enrolled and randomly assigned to either gemcitabine (1,000 mg/m2, days 1 and 4), epirubicin (90 mg/m2, day 1), and paclitaxel (175 mg/m2, day 1) or FU (500 mg/m2, day 1), epirubicin (90 mg/m2, day 1), and cyclophosphamide (500 mg/m2, day 1). Both regimens were administered every 21 days for a maximum of eight cycles. Results Between October 1999 and November 2002, 259 patients (GET, n = 124; FEC, n = 135) were enrolled. Baseline characteristics were well balanced across treatment arms. After a median of 20.4 months of follow-up, median TtPD was 9.1 months and 9.0 months in the GET and FEC arms, respectively (P = .557). The ORR was 62.3% in the GET arm (n = 114) and 51.2% in the FEC arm (n = 129; P = .093). Grade 3 and 4 toxicities, including neutropenia, thrombocytopenia, anemia, stomatitis, neurosensory toxicity, and allergy, occurred significantly more often in the GET arm. Conclusion No significant differences in terms of TtPD and ORR were observed between the two treatment arms. Treatment-related toxicity was higher in the GET arm.
Collapse
Affiliation(s)
- Christoph Zielinski
- Department of Medicine I, University Hospital, 18-20 Waehringer Guertel, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Fifty percent of breast cancers occur after the age of 65 years and 25% occur after the age of 75 years. Encountering a breast cancer in an older woman is frequent. After years of dearth of data specific to the elderly, some evidence is beginning to accumulate concerning breast cancer in the older woman. Recent data from mammography studies confirm its effective in women with 10 years or more of life expectancy (perhaps even 5 years). Epidemiologic and randomized studies demonstrate that a proper surgery and adjuvant treatment can decrease relapse and improve survival in patients older than 80 years. Radiation therapy studies show a decrease in local relapse even in patients older than 70 years. Adjuvant hormonal therapy has essentially the same effectiveness as in younger women. Chemotherapy has a role in patients older than 70 years. Consensus statements, such as the St. Gallen consensus, have dropped the age limit of 70 years from their recommendations. Comorbidity and life expectancy should be taken into account for proper selection of adjuvant treatment. The treatment of metastatic breast cancer has evolved significantly with the introduction of aromatase inhibitors, new chemotherapeutic agents, and targeted biologic agents. New chemotherapeutic agents are as effective as single agents compared to older and more toxic drug combinations. The cumulative result of the introduction of these new agents, at a population level, is a 7.5-month increase in the median survival time of patients with metastatic breast cancer over the past decade.
Collapse
|