201
|
P53/Rb inhibition induces metastatic adrenocortical carcinomas in a preclinical transgenic model. Oncogene 2017; 36:4445-4456. [PMID: 28368424 DOI: 10.1038/onc.2017.54] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/20/2016] [Accepted: 02/04/2017] [Indexed: 12/14/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare cancer with poor prognosis. Pan-genomic analyses identified p53/Rb and WNT/β-catenin signaling pathways as main contributors to the disease. However, isolated β-catenin constitutive activation failed to induce malignant progression in mouse adrenocortical tumors. Therefore, there still was a need for a relevant animal model to study ACC pathogenesis and to test new therapeutic approaches. Here, we have developed a transgenic mice model with adrenocortical specific expression of SV40 large T-antigen (AdTAg mice), to test the oncogenic potential of p53/Rb inhibition in the adrenal gland. All AdTAg mice develop large adrenal carcinomas that eventually metastasize to the liver and lungs, resulting in decreased overall survival. Consistent with ACC in patients, adrenal tumors in AdTAg mice autonomously produce large amounts of glucocorticoids and spontaneously activate WNT/β-catenin signaling pathway during malignant progression. We show that this activation is associated with downregulation of secreted frizzled related proteins (Sfrp) and Znrf3 that act as inhibitors of the WNT signaling. We also show that mTORC1 pathway activation is an early event during neoplasia expansion and further demonstrate that mTORC1 pathway is activated in ACC patients. Preclinical inhibition of mTORC1 activity induces a marked reduction in tumor size, associated with induction of apoptosis and inhibition of proliferation that results in normalization of corticosterone plasma levels in AdTAg mice. Altogether, these data establish AdTAg mice as the first preclinical model for metastatic ACC.
Collapse
|
202
|
Fernandes JC, Rodrigues Alves APN, Machado-Neto JA, Scopim-Ribeiro R, Fenerich BA, da Silva FB, Simões BP, Rego EM, Traina F. IRS1/β-Catenin Axis Is Activated and Induces MYC Expression in Acute Lymphoblastic Leukemia Cells. J Cell Biochem 2017; 118:1774-1781. [DOI: 10.1002/jcb.25845] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/14/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Jaqueline Cristina Fernandes
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | | | - João Agostinho Machado-Neto
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Renata Scopim-Ribeiro
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Bruna Alves Fenerich
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Fernanda Borges da Silva
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Belinda Pinto Simões
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Eduardo Magalhães Rego
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Fabiola Traina
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| |
Collapse
|
203
|
Ruggiero C, Doghman-Bouguerra M, Sbiera S, Sbiera I, Parsons M, Ragazzon B, Morin A, Robidel E, Favier J, Bertherat J, Fassnacht M, Lalli E. Dosage-dependent regulation of VAV2 expression by steroidogenic factor-1 drives adrenocortical carcinoma cell invasion. Sci Signal 2017; 10:eaal2464. [PMID: 28270555 DOI: 10.1126/scisignal.aal2464] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with a dismal prognosis. Genomic studies have enabled progress in our understanding of the molecular bases of ACC, but factors that influence its prognosis are lacking. Amplification of the gene encoding the transcription factor steroidogenic factor-1 (SF-1; also known as NR5A1) is one of the genetic alterations common in ACC. We identified a transcriptional regulatory mechanism involving increased abundance of VAV2, a guanine nucleotide exchange factor for small GTPases that control the cytoskeleton, driven by increased expression of the gene encoding SF-1 in ACC. Manipulating SF-1 and VAV2 abundance in cultured ACC cells revealed that VAV2 was a critical factor for SF-1-induced cytoskeletal remodeling and invasion in culture (Matrigel) and in vivo (chicken chorioallantoic membrane) models. Analysis of ACC patient cohorts indicated that greater VAV2 abundance robustly correlated with poor prognosis in ACC patients. Because VAV2 is a druggable target, our findings suggest that blocking VAV2 may be a new therapeutic approach to inhibit metastatic progression in ACC patients.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France
- CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| | - Mabrouka Doghman-Bouguerra
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France
- CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Iuliu Sbiera
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, U.K
| | - Bruno Ragazzon
- Inserm, U1016, Institut Cochin, 75014 Paris, France
- CNRS UMR8104, 75014 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
| | - Aurélie Morin
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
- Inserm, UMR970, Paris Cardiovascular Research Centre, 75015 Paris, France
| | - Estelle Robidel
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
- Inserm, UMR970, Paris Cardiovascular Research Centre, 75015 Paris, France
| | - Judith Favier
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
- Inserm, UMR970, Paris Cardiovascular Research Centre, 75015 Paris, France
| | - Jérôme Bertherat
- Inserm, U1016, Institut Cochin, 75014 Paris, France
- CNRS UMR8104, 75014 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
| | - Martin Fassnacht
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| | - Enzo Lalli
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France.
- CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| |
Collapse
|
204
|
Gaujoux S, Mihai R. European Society of Endocrine Surgeons (ESES) and European Network for the Study of Adrenal Tumours (ENSAT) recommendations for the surgical management of adrenocortical carcinoma. Br J Surg 2017; 104:358-376. [PMID: 28199015 DOI: 10.1002/bjs.10414] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/10/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Radical surgery provides the best chance of cure for adrenocortical carcinoma (ACC), but perioperative surgical care for these patients is yet to be standardized. METHODS A working group appointed jointly by ENSAT and ESES used Delphi methodology to produce evidence-based recommendations for the perioperative surgical care of patients with ACC. Papers were retrieved from electronic databases. Evidence and recommendations were classified according to the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) system, and were discussed until consensus was reached within the group. RESULTS Twenty-five recommendations for the perioperative surgical care of patients with ACC were formulated. The quality of evidence is low owing to the rarity of the disease and the lack of prospective surgical trials. Multi-institutional prospective cohort studies and prospective RCTs are urgently needed and should be strongly encouraged. CONCLUSION The present evidence-based recommendations provide comprehensive advice on the optimal perioperative care for patients undergoing surgery for ACC.
Collapse
Affiliation(s)
- S Gaujoux
- Department of Digestive and Endocrine Surgery, Cochin Hospital, Assistance Publique - Hôpitaux de Paris (APHP), Paris, France
- Faculté de Médecine Paris Descartes, Université Paris Descartes, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unit 1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut Cochin, Paris, France
| | - R Mihai
- Churchill Cancer Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| | | |
Collapse
|
205
|
Stigliano A, Cerquetti L, Lardo P, Petrangeli E, Toscano V. New insights and future perspectives in the therapeutic strategy of adrenocortical carcinoma (Review). Oncol Rep 2017; 37:1301-1311. [PMID: 28184938 DOI: 10.3892/or.2017.5427] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/20/2016] [Indexed: 11/06/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with an incidence ranging from 0.7 to 2.0 cases/million people per year. Hypercortisolism represents the most common clinical presentation in many patients although, less frequently, some ACC secreting androgens and estrogens are even more pathognomonic compared to cortisol secretion. Currently, radical surgery, when feasible, is still the only curative therapy. Mitotane, an adrenolytic drug, is used in the adjuvant setting and in combination with chemotherapy drugs in metastatic disease. The use of radiotherapy remains controversial, being indicated only in selected cases. New targeted therapies, such as insulin growth factor-1 (IGF-1), mammalian-target of rapamycin (m-TOR), vascular endothelial growth factor (VEGF) inhibitors and others, have recently been investigated with disappointing clinical results. The partial effectiveness of current treatments mandates the need for new therapeutic strategies against this tumor.
Collapse
Affiliation(s)
- Antonio Stigliano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Lidia Cerquetti
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Pina Lardo
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Elisa Petrangeli
- CNR, Institute of Molecular Biology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Vincenzo Toscano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
206
|
Li H, Batth IS, Qu X, Xu L, Song N, Wang R, Liu Y. IGF-IR signaling in epithelial to mesenchymal transition and targeting IGF-IR therapy: overview and new insights. Mol Cancer 2017; 16:6. [PMID: 28137302 PMCID: PMC5282886 DOI: 10.1186/s12943-016-0576-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023] Open
Abstract
The insulin-like growth factor-I (IGF-I) signaling induces epithelial to mesenchymal transition (EMT) program and contributes to metastasis and drug resistance in several subtypes of tumors. In preclinical studies, targeting of the insulin-like growth factor-I receptor (IGF-IR) showed promising anti-tumor effects. Unfortunately, high expectations for anti-IGF-IR therapy encountered challenge and disappointment in numerous clinical trials. This review summarizes the regulation of EMT by IGF-I/IGF-IR signaling pathway and drug resistance mechanisms of targeting IGF-IR therapy. Most importantly, we address several factors in the regulation of IGF-I/IGF-IR-associated EMT progression that may be potential predictive biomarkers in targeted therapy.
Collapse
Affiliation(s)
- Heming Li
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China.,Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Izhar Singh Batth
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Ling Xu
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Na Song
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Ruoyu Wang
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.
| | - Yunpeng Liu
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China.
| |
Collapse
|
207
|
Leighl NB, Rizvi NA, de Lima LG, Arpornwirat W, Rudin CM, Chiappori AA, Ahn MJ, Chow LQM, Bazhenova L, Dechaphunkul A, Sunpaweravong P, Eaton K, Chen J, Medley S, Poondru S, Singh M, Steinberg J, Juergens RA, Gadgeel SM. Phase 2 Study of Erlotinib in Combination With Linsitinib (OSI-906) or Placebo in Chemotherapy-Naive Patients With Non-Small-Cell Lung Cancer and Activating Epidermal Growth Factor Receptor Mutations. Clin Lung Cancer 2017; 18:34-42.e2. [PMID: 27686971 PMCID: PMC5474312 DOI: 10.1016/j.cllc.2016.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 07/29/2016] [Indexed: 12/23/2022]
Abstract
INTRODUCTION First-line epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor treatment of advanced non-small-cell lung cancer with EGFR-activating mutations improves outcomes compared with chemotherapy, but resistance develops in most patients. Compensatory signaling through type 1 insulin-like growth factor 1 receptor (IGF-1R) may contribute to resistance; dual blockade of IGF-1R and EGFR may improve outcomes. PATIENTS AND METHODS We performed a randomized, double-blind, placebo-controlled phase II study of linsitinib, a dual IGF-1R and insulin receptor tyrosine kinase inhibitor, plus erlotinib versus placebo plus erlotinib in chemotherapy-naive patients with EGFR-mutation positive, advanced non-small-cell lung cancer. Patients received linsitinib 150 mg twice daily or placebo plus erlotinib 150 mg once daily on continuous 21-day cycles. The primary end point was progression-free survival. RESULTS After randomization of 88 patients (44 each arm), the trial was unblinded early owing to inferiority in the linsitinib arm. The median progression-free survival for the linsitinib versus the placebo group was 8.4 months versus 12.4 months (hazard ratio, 1.37; P = .29). Overall response rate (47.7% vs. 75.0%; P = .02) and disease control rate (77.3% vs. 95.5%; P = .03) were also inferior. Whereas most adverse events were ≤ grade 2, linsitinib plus erlotinib was associated with increased adverse events that led to decreased erlotinib exposure (median days, 228 vs. 305). No drug-drug interaction was suggested by pharmacokinetic and pharmacodynamic results. CONCLUSION Adding linsitinib to erlotinib resulted in inferior outcomes compared with erlotinib alone. Further understanding of the signaling pathways and a biomarker that can predict efficacy is needed prior to further clinical development of IGF-1R inhibitors in lung cancer.
Collapse
Affiliation(s)
- Natasha B Leighl
- Cancer Clinical Research Unit, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Naiyer A Rizvi
- Department of Hematology and Oncology, Columbia University Medical Center, New York, NY
| | - Lopes Gilberto de Lima
- Oncoclinicas Group, Hcor Onco, Sao Paulo, SP, Brazil; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | | | | | | | - Myung-Ju Ahn
- Department of Hematology and Oncology, Sungkyunkwan University, Seoul, South Korea
| | | | - Lyudmila Bazhenova
- Department of Clinical Oncology, School of Medicine, UC San Diego Moores Cancer Center, San Diego, CA
| | - Arunee Dechaphunkul
- Departments of Pathology, Hematology, and Surgery, Prince of Songkla University, Songkhla, Thailand
| | | | - Keith Eaton
- Prince of Songkla University, Songkhla, Thailand; Medical Oncology at University of Washington School of Medicine, Seattle Cancer Care Alliance, Seattle, WA
| | | | | | | | | | | | - Rosalyn A Juergens
- Juravinski Cancer Centre, Department of Oncology, McMaster University, Hamilton, ON, Canada.
| | - Shirish M Gadgeel
- Karmanos Cancer Institute, Wayne State University, Detroit, MI; Hematology-Oncology, Wayne State University, Detroit, MI
| |
Collapse
|
208
|
Abstract
The recently available genomic sequencing techniques have led to breakthroughs in understanding of the underlying genetic mechanisms in adrenocortical tumours. Disease-causing mutations have been described for aldosterone-producing adenomas, cortisol-producing adenomas and adrenocortical carcinomas. Further, knowledge gained from transcriptome analyses and methylation arrays has provided new insights into the development of these tumours. Elucidation of the genomic landscape of adrenocortical tumours and improved techniques may in the future be useful for early diagnosis through the detection of mutated DNA in the circulation. Moreover, compounds that bind specifically to altered proteins may be used as screening targets or therapeutic agents. Regulation of cortisol release by interaction with an altered subunit in adenylate cyclase may be more complex, but may provide a new option for regulating steroid release. Information about derangements in adrenocortical carcinoma is already helpful for determining patient prognosis. With further knowledge, we may be able to identify novel biomarkers that effectively and noninvasively help in differentiating between benign and malignant disease. It is clear that the next few years will provide much novel information that hopefully will aid in the treatment of patients with adrenocortical tumours.
Collapse
Affiliation(s)
- T Åkerström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - T Carling
- Endocrine Research Unit, Yale University, New Haven, CT, USA
| | - F Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - P Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
209
|
Insulin-IGF signaling affects cell transformation in the BALB/c 3T3 cell model. Sci Rep 2016; 6:37120. [PMID: 27849005 PMCID: PMC5111065 DOI: 10.1038/srep37120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022] Open
Abstract
The increased cancer mortality of diabetes type 2 patients is most likely an evidence of the tight connection between tumor development and energy metabolism. A major focus of today's research is still the identification of key proteins of both diseases and the development of corresponding inhibitors. In this study we combined the two-stage BALB/c-3T3 cell transformation assay (BALB-CTA) with the IR/IGF-1R inhibitor OSI-906 (linsitinib) and analyzed alterations in protein activity and energy parameters in non-transformed as well as transformed cells. OSI-906 successfully inhibited the phosphorylation of IR/IGF-1R and decreased cell growth in non-transformed cells. In the BALB-CTA, a permanent treatment with OSI-906 reduced cellular transformation dose-dependently, whereas a temporary treatment gave evidence for a preventive effect in the promotion phase. Furthermore, even though several key proteins were affected, it was possible to show that the phosphorylation of GSK3, Erk 1/2 and the S6 protein are not crucial for the cell foci reducing effect of OSI-906. Taken together, the BALB-CTA confirmed results of OSI-906 from animal studies and enhanced the knowledge of its mode of action. Therefore, the BALB-CTA offers the opportunity to analyze alterations in the transformation process more precisely and will be helpful to identify effective cancer treatments.
Collapse
|
210
|
van Beijnum JR, Pieters W, Nowak-Sliwinska P, Griffioen AW. Insulin-like growth factor axis targeting in cancer and tumour angiogenesis - the missing link. Biol Rev Camb Philos Soc 2016; 92:1755-1768. [PMID: 27779364 DOI: 10.1111/brv.12306] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/15/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
Numerous molecular players in the process of tumour angiogenesis have been shown to offer potential for therapeutic targeting. Initially denoted to be involved in malignant transformation and tumour progression, the insulin-like growth factor (IGF) signalling axis has been subject to therapeutic interference, albeit with limited clinical success. More recently, IGFs and their receptors have received attention for their contribution to tumour angiogenesis, which offers novel therapeutic opportunities. Here we review the contribution of this signalling axis to tumour angiogenesis, the mechanisms of resistance to therapy and the interplay with other pro-angiogenic pathways, to offer insight in the renewed interest in the application of IGF axis targeting agents in anti-cancer combination therapies.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Wietske Pieters
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva (UNIGE), Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Arjan W Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| |
Collapse
|
211
|
Sclafani F, Kim TY, Cunningham D, Kim TW, Tabernero J, Schmoll HJ, Roh JK, Kim SY, Park YS, Guren TK, Hawkes E, Clarke SJ, Ferry D, Frodin JE, Ayers M, Nebozhyn M, Peckitt C, Loboda A, Watkins DJ. Dalotuzumab in chemorefractory KRAS exon 2 mutant colorectal cancer: Results from a randomised phase II/III trial. Int J Cancer 2016; 140:431-439. [PMID: 27681944 DOI: 10.1002/ijc.30453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/30/2022]
Abstract
Limited data are available on the efficacy of anti-IGF-1R agents in KRAS mutant colorectal cancer (CRC). We analysed the outcome of 69 chemorefractory, KRAS exon 2 mutant CRC patients who were enrolled in a double-blind, randomised, phase II/III study of irinotecan and cetuximab plus dalotuzumab 10 mg/kg once weekly (arm A), dalotuzumab 7.5 mg/kg every second week (arm B) or placebo (arm C). Objective response rate (5.6% vs. 3.1% vs. 4.8%), median progression-free survival (2.7 vs. 2.6 vs. 1.4 months) and overall survival (7.8 vs. 10.3 vs. 7.8 months) were not statistically significantly different between treatment groups. Most common grade ≥3 treatment-related toxicities included neutropenia, diarrhoea, hyperglycaemia, fatigue and dermatitis acneiform. Expression of IGF-1R, IGF-1, IGF-2 and EREG by quantitative real-time polymerase chain reaction was assessed in 351 patients from the same study with available data on KRAS exon 2 mutational status. Median cycle threshold values for all biomarkers were significantly lower (i.e., higher expression, p < 0.05) among patients with KRAS wild-type compared to those with KRAS exon 2 mutant tumours. No significant changes were found according to location of the primary tumour with only a trend towards lower expression of IGF-1 in colon compared to rectal cancers (p = 0.06). Albeit limited by the small sample size, this study does not appear to support a potential role for anti-IGF-1R agents in KRAS exon 2 mutant CRC. Data on IGF-1R, IGF-1 and IGF-2 expression here reported may be useful for patient stratification in future trials with inhibitors of the IGF pathway.
Collapse
Affiliation(s)
- Francesco Sclafani
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Tae Y Kim
- Seoul National University College of Medicine, Seoul, Korea
| | - David Cunningham
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Tae W Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Hans J Schmoll
- Department of Internal Medicine, University Clinic Halle (Saale), Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jae K Roh
- College of Medicine, Yonsey Cancer Center, Yonsey University, Seoul, Korea
| | - Sun Y Kim
- Center for Colorectal Cancer, National Cancer Center, Seoul, Korea
| | - Young S Park
- Department of Medicine, Division of Hematology/Oncology, Samsung Medical Center, Seoul, Korea
| | - Tormod K Guren
- Department of Oncology and K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Eliza Hawkes
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Stephen J Clarke
- Concord Repatriation General Hospital, Concord, Sydney, Australia
| | - David Ferry
- New Cross Hospital, Wolverhamptom, United Kingdom
| | | | | | | | - Clare Peckitt
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | | | - David J Watkins
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| |
Collapse
|
212
|
Jung S, Nagy Z, Fassnacht M, Zambetti G, Weiss M, Reincke M, Igaz P, Beuschlein F, Hantel C. Preclinical progress and first translational steps for a liposomal chemotherapy protocol against adrenocortical carcinoma. Endocr Relat Cancer 2016; 23:825-37. [PMID: 27550961 DOI: 10.1530/erc-16-0249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 02/02/2023]
Abstract
Systemic therapy of adrenocortical carcinoma (ACC) is limited by heterogeneous tumor response and adverse effects. Recently, we demonstrated anti-tumor activity of LEDP-M (etoposide, liposomal doxorubicin, liposomal cisplatin, mitotane), a liposomal variant of EDP-M (etoposide, doxorubicin, cisplatin, mitotane). To improve the therapeutic efficacy and off-target profiles of the clinical gold standard EDP-M, we investigated liposomal EDP-M regimens in different preclinical settings and in a small number of ACC patients with very advanced disease. Short- and long-term experiments were performed on two ACC models (SW-13 and SJ-ACC3) in vivo We evaluated the anti-tumoral effects and off-target profiles of EDP-M, LEDP-M and a novel regimen L(l)EDP-M including liposomal etoposide. Furthermore, the role of plasma microRNA-210 as a therapeutic biomarker and first clinical data were assessed. Classical and liposomal protocols revealed anti-proliferative efficacy against SW-13 (EDP-M P < 0.01; LEDP-M: P < 0.001; L(l)EDP-M: P < 0.001 vs controls), whereas in SJ-ACC3, only EDP-M (P < 0.05 vs controls) was slightly effective. Long-term experiments in SW-13 demonstrated anti-tumor efficacy for all treatment schemes (EDP-M: P < 0.01, LEDP-M: P < 0.05, L(l)EDP-M P < 0.001 vs controls). The analysis of pre-defined criteria leading to study termination revealed significant differences for control (P < 0.0001) and EDP-M (P = 0.003) compared to L(l)EDP-M treatment. Raising its potential for therapy monitoring, we detected elevated levels of circulating microRNA-210 in SW-13 after LEDP-M treatment (P < 0.05). In contrast, no comparable effects were detectable for SJ-ACC3. However, overall histological evaluation demonstrated improved off-target profiles following liposomal regimens. The first clinical data indicate improved tolerability of liposomal EDP-M, thus confirming our results. In summary, liposomal EDP-M regimens represent promising treatment options to improve clinical treatment of ACC.
Collapse
Affiliation(s)
- Sara Jung
- Endocrine Research UnitMedizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Germany
| | - Zoltan Nagy
- 2nd Department of MedicineSemmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Martin Fassnacht
- Department of Internal Medicine IDivision of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany Comprehensive Cancer Center MainfrankenUniversity of Würzburg, Würzburg, Germany
| | - Gerard Zambetti
- Department of PathologySt Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Max Weiss
- Institute of PathologyLudwig-Maximilians-University, Munich, Germany
| | - Martin Reincke
- Endocrine Research UnitMedizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Germany
| | - Peter Igaz
- 2nd Department of MedicineSemmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Felix Beuschlein
- Endocrine Research UnitMedizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Germany
| | - Constanze Hantel
- Endocrine Research UnitMedizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
213
|
Clinical studies in humans targeting the various components of the IGF system show lack of efficacy in the treatment of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 772:105-122. [PMID: 28528684 DOI: 10.1016/j.mrrev.2016.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 01/28/2023]
Abstract
The insulin-like growth factors (IGFs) system regulates cell growth, differentiation and energy metabolism and plays crucial role in the regulation of key aspects of tumor biology, such as cancer cell growth, survival, transformation and invasion. The current focus for cancer therapeutic approaches have shifted from the conventional treatments towards the targeted therapies and the IGF system has gained a great interest as anti-cancer therapy. The proliferative, anti-apoptotic and transformation effects of IGFs are mainly triggered by the ligation of the type I IGF receptor (IGF-IR). Thus, aiming at developing novel and effective cancer therapies, different strategies have been employed to target IGF system in human malignancies, including but not limited to ligand or receptor neutralizing antibodies and IGF-IR signaling inhibitors. In this review, we have focused on the clinical studies that have been conducted targeting the various components of the IGF system for the treatment of different types of cancer, providing a description and the challenges of each targeting strategy and the degree of success.
Collapse
|
214
|
Goldman JW, Mendenhall MA, Rettinger SR. Hyperglycemia Associated With Targeted Oncologic Treatment: Mechanisms and Management. Oncologist 2016; 21:1326-1336. [PMID: 27473045 DOI: 10.1634/theoncologist.2015-0519] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/09/2016] [Indexed: 12/15/2022] Open
Abstract
: Molecularly targeted cancer therapy has rapidly changed the landscape of oncologic care, often improving patients' prognosis without causing as substantial a quality-of-life decrement as cytotoxic chemotherapy does. Nevertheless, targeted agents can cause side effects that may be less familiar to medical oncologists and that require the attention and expertise of subspecialists. In this review, we focus on hyperglycemia, which can occur with use of new anticancer agents that interact with cell proliferation pathways. Key mediators of these pathways include the tyrosine kinase receptors insulin growth factor receptor 1 (IGF-1R) and epidermal growth factor receptor (EGFR), as well as intracellular signaling molecules phosphatidylinositol 3-kinase (PI3K), AKT, and mammalian target of rapamycin (mTOR). We summarize available information on hyperglycemia associated with agents that inhibit these molecules within the larger context of adverse event profiles. The highest incidence of hyperglycemia is observed with inhibition of IGF-1R or mTOR, and although the incidence is lower with PI3K, AKT, and EGFR inhibitors, hyperglycemia is still a common adverse event. Given the interrelationships between the IGF-1R and cell proliferation pathways, it is important for oncologists to understand the etiology of hyperglycemia caused by anticancer agents that target those pathways. We also discuss monitoring and management approaches for treatment-related hyperglycemia for some of these agents, with a focus on our experience during the clinical development of the EGFR inhibitor rociletinib. IMPLICATIONS FOR PRACTICE Treatment-related hyperglycemia is associated with several anticancer agents. Many cancer patients may also have preexisting or undiagnosed diabetes or glucose intolerance. Screening can identify patients at risk for hyperglycemia before treatment with these agents. Proper monitoring and management of symptoms, including lifestyle changes and pharmacologic intervention, may allow patients to continue benefiting from use of anticancer agents.
Collapse
Affiliation(s)
- Jonathan W Goldman
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Melody A Mendenhall
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Sarah R Rettinger
- Endocrinology Medical Group of Orange County, Inc., Orange, California, USA
| |
Collapse
|
215
|
Beuschlein F, Jakoby J, Mentz S, Zambetti G, Jung S, Reincke M, Süss R, Hantel C. IGF1-R inhibition and liposomal doxorubicin: Progress in preclinical evaluation for the treatment of adrenocortical carcinoma. Mol Cell Endocrinol 2016; 428:82-8. [PMID: 26994514 DOI: 10.1016/j.mce.2016.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 11/17/2022]
Abstract
Adrenocortical carcinoma (ACC) is a tumor with poor prognosis and limited therapeutic options. Therefore, in addition to multi-chemotherapeutic regimens IGF-1 receptor (IGF-1R) targeting approaches have been evaluated including immunoliposomal (IL) preparations utilizing an IGF-1R inhibiting antibody. In the current study, we extended our experiments by long-term treatment regimens in the classical adrenocortical NCIH295R xenograft model as well as by short-term experiments in two novel xenograft models, which all displayed different levels of IGF-1R and IGF-2 expression. Interestingly, these experiments reveal sub-group dependent differences in therapeutic outcome, reflecting clinical observations and indicate, thus, that implementation of this panel of tumor models might be helpful for clinical translation of novel therapeutic regimens in the future.
Collapse
Affiliation(s)
- Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Judith Jakoby
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Susanne Mentz
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gerard Zambetti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sara Jung
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Reincke
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Regine Süss
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Constanze Hantel
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
216
|
Abstract
PURPOSE OF REVIEW Adrenocortical carcinoma is a rare cancer, but one that carries a poor prognosis due to its aggressive nature and unresponsiveness to conventional chemotherapeutic strategies. Over the past 12 years, there has been renewed interest in developing new therapies for this cancer, including identifying key signaling nodes responsible for cell proliferation. RECENT FINDINGS Clinical trials of tyrosine kinase inhibitors as monotherapy have generally been disappointing, although the identification of exceptional responders may lead to the identification of targeted therapies that may produce responses in subsets of patients. Agents targeted to the Wnt signaling pathway, a known player in adrenal carcinogenesis, have been developed, although they have not yet been used specifically for adrenal cancer. There is current excitement about inhibitors of acetyl-coA cholesterol acetyl transferase 1, an enzyme required for intracellular cholesterol handling, although trials are still underway. Tools to target other proteins such as Steroidogenic Factor 1 and mechanistic target of rapamycin have been developed and are moving towards clinical application. SUMMARY Progress is being made in the fight against adrenocortical carcinoma with the identification of new therapeutic targets and new means by which to attack them. Continued improvement in the prognosis for patients with adrenal cancer is expected as this research continues.
Collapse
Affiliation(s)
- Bhavana Konda
- aDivision of OncologybDivision of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | | |
Collapse
|
217
|
Peixoto Lira RC, Fedatto PF, Marco Antonio DS, Leal LF, Martinelli CE, de Castro M, Tucci S, Neder L, Ramalho L, Seidinger AL, Cardinalli I, Mastellaro MJ, Yunes JA, Brandalise SR, Tone LG, Rauber Antonini SR, Scrideli CA. IGF2 and IGF1R in pediatric adrenocortical tumors: roles in metastasis and steroidogenesis. Endocr Relat Cancer 2016; 23:481-93. [PMID: 27185872 DOI: 10.1530/erc-15-0426] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/16/2016] [Indexed: 01/01/2023]
Abstract
Deregulation of the IGF system observed in human tumors indicates a role in malignant cell transformation and in tumor cell proliferation. Although overexpression of the IGF2 and IGF1R genes was described in adrenocortical tumors (ACTs), few studies reported their profiles in pediatric ACTs. In this study, the IGF2 and IGF1R expression was evaluated by RT-qPCR according to the patient's clinical/pathological features in 60 pediatric ACT samples, and IGF1R protein was investigated in 45 samples by immunohistochemistry (IHC). Whole transcriptome and functional assays were conducted after IGF1R inhibition with OSI-906 in NCI-H295A cell line. Significant IGF2 overexpression was found in tumor samples when compared with non-neoplastic samples (P<0.001), significantly higher levels of IGF1R in patients with relapse/metastasis (P=0.031) and moderate/strong IGF1R immunostaining in 62.2% of ACTs, but no other relationship with patient survival and clinical/pathological features was observed. OSI-906 treatment downregulated genes associated with MAPK activity, induced limited reduction of cell viability and increased the apoptosis rate. After 24h, the treatment also decreased the expression of genes related to the steroid biosynthetic process, the protein levels of the steroidogenic acute regulatory protein (STAR), and androgen secretion in cell medium, supporting the role of IGF1R in steroidogenesis of adrenocortical carcinoma cells. Our data showed that the IGF1R overexpression could be indicative of aggressive ACTs in children. However, in vitro treatments with high concentrations of OSI-906 (>1μM) showed limited reduction of cell viability, suggesting that OSI-906 alone could not be a suitable therapy to abolish carcinoma cell growth.
Collapse
Affiliation(s)
- Régia Caroline Peixoto Lira
- Department of PediatricsRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Paola Fernanda Fedatto
- Department of PediatricsRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | | | - Letícia Ferro Leal
- Department of PediatricsRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Carlos Eduardo Martinelli
- Department of PediatricsRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Margaret de Castro
- Department of Internal MedicineRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Silvio Tucci
- Department of SurgeryRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Luciano Neder
- Department of PathologyRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Leandra Ramalho
- Department of PathologyRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Ana Luiza Seidinger
- Boldrini Children CenterState University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Izilda Cardinalli
- Boldrini Children CenterState University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Maria José Mastellaro
- Boldrini Children CenterState University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - José Andres Yunes
- Boldrini Children CenterState University of Campinas (UNICAMP), Campinas, São Paulo, Brazil State University of Campinas (UNICAMP)CampinasSão Paulo, Brazil
| | - Silvia Regina Brandalise
- Boldrini Children CenterState University of Campinas (UNICAMP), Campinas, São Paulo, Brazil State University of Campinas (UNICAMP)CampinasSão Paulo, Brazil
| | - Luiz Gonzaga Tone
- Department of PediatricsRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | | | - Carlos Alberto Scrideli
- Department of PediatricsRibeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| |
Collapse
|
218
|
de Lint K, Poell JB, Soueidan H, Jastrzebski K, Vidal Rodriguez J, Lieftink C, Wessels LF, Beijersbergen RL. Sensitizing Triple-Negative Breast Cancer to PI3K Inhibition by Cotargeting IGF1R. Mol Cancer Ther 2016; 15:1545-56. [DOI: 10.1158/1535-7163.mct-15-0865] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/26/2016] [Indexed: 11/16/2022]
|
219
|
Batson S, Greenall G, Hudson P. Review of the Reporting of Survival Analyses within Randomised Controlled Trials and the Implications for Meta-Analysis. PLoS One 2016; 11:e0154870. [PMID: 27149107 PMCID: PMC4858202 DOI: 10.1371/journal.pone.0154870] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/20/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Meta-analysis is a growing approach to evidence synthesis and network meta-analysis in particular represents an important and developing method within Health Technology Assessment (HTA). Meta-analysis of survival data is usually performed using the individual summary statistic-the hazard ratio (HR) from each randomised controlled trial (RCT). OBJECTIVES The objectives of this study are to: (i) review the methods and reporting of survival analyses in oncology RCTs; and (ii) assess the suitability and relevance of survival data reported in RCTs for inclusion into meta-analysis. METHODS Five oncology journals were searched to identify Phase III RCTs published between April and July 2015. Eligible studies included those that analysed a survival outcome. RESULTS Thirty-two RCTs reporting survival outcomes in cancer populations were identified. None of the publications reported details relating to a strategy for statistical model building, the goodness of fit of the final model, or final model validation for the analysis of survival outcomes. The majority of studies (88%) reported the use of Cox proportional hazards (PH) regression to analyse survival endpoints. However, most publications failed to report the validation of the statistical models in terms of the PH assumption. CONCLUSIONS This review highlights deficiencies in terms of reporting the methods and validity of survival analyses within oncology RCTs. We support previous recommendations to encourage authors to improve the reporting of survival analyses in journal publications. We also recommend that the final choice of a statistical model for survival should be informed by goodness of model fit to a given dataset, and that model assumptions are validated. The failure of trial investigators and statisticians to investigate the PH for RCT survival data is likely to result in clinical decisions based on inappropriate methods. The development of alternative approaches for the meta-analysis of survival outcomes when the PH assumption is implausible is required if valid clinical decisions are to be made.
Collapse
|
220
|
Drelon C, Berthon A, Mathieu M, Ragazzon B, Kuick R, Tabbal H, Septier A, Rodriguez S, Batisse-Lignier M, Sahut-Barnola I, Dumontet T, Pointud JC, Lefrançois-Martinez AM, Baron S, Giordano TJ, Bertherat J, Martinez A, Val P. EZH2 is overexpressed in adrenocortical carcinoma and is associated with disease progression. Hum Mol Genet 2016; 25:2789-2800. [PMID: 27149985 DOI: 10.1093/hmg/ddw136] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 12/29/2022] Open
Abstract
Adrenal Cortex Carcinoma (ACC) is an aggressive tumour with poor prognosis. Common alterations in patients include constitutive WNT/β-catenin signalling and overexpression of the growth factor IGF2. However, the combination of both alterations in transgenic mice is not sufficient to trigger malignant tumour progression, suggesting that other alterations are required to allow development of carcinomas. Here, we have conducted a study of publicly available gene expression data from three cohorts of ACC patients to identify relevant alterations. Our data show that the histone methyltransferase EZH2 is overexpressed in ACC in the three cohorts. This overexpression is the result of deregulated P53/RB/E2F pathway activity and is associated with increased proliferation and poorer prognosis in patients. Inhibition of EZH2 by RNA interference or pharmacological treatment with DZNep inhibits cellular growth, wound healing and clonogenic growth and induces apoptosis of H295R cells in culture. Further growth inhibition is obtained when DZNep is combined with mitotane, the gold-standard treatment for ACC. Altogether, these observations suggest that overexpression of EZH2 is associated with aggressive progression and may constitute an interesting therapeutic target in the context of ACC.
Collapse
Affiliation(s)
- Coralie Drelon
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Annabel Berthon
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France.,Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892-1103, USA
| | - Mickael Mathieu
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Bruno Ragazzon
- Inserm U1016, CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Rork Kuick
- Department of Biostatistics, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Houda Tabbal
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Amandine Septier
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Stéphanie Rodriguez
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Marie Batisse-Lignier
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France.,Centre Hospitalier Universitaire, Service d'Endocrinologie, Faculté de Médecine, F- 63000 Clermont-Ferrand, France
| | - Isabelle Sahut-Barnola
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Typhanie Dumontet
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | | | | | - Silvère Baron
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Thomas J Giordano
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Jérôme Bertherat
- Inserm U1016, CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Antoine Martinez
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| | - Pierre Val
- CNRS, UMR6293, GReD, Inserm U1103, Clermont Université, F-63001 Clermont-Ferrand, France
| |
Collapse
|
221
|
Sun H, Lin DC, Cao Q, Guo X, Marijon H, Zhao Z, Gery S, Xu L, Yang H, Pang B, Lee VKM, Lim HJ, Doan N, Said JW, Chu P, Mayakonda A, Thomas T, Forscher C, Baloglu E, Shacham S, Rajalingam R, Koeffler HP. CRM1 Inhibition Promotes Cytotoxicity in Ewing Sarcoma Cells by Repressing EWS-FLI1-Dependent IGF-1 Signaling. Cancer Res 2016; 76:2687-97. [PMID: 26956669 DOI: 10.1158/0008-5472.can-15-1572] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 02/19/2016] [Indexed: 11/16/2022]
Abstract
Ewing sarcoma (EWS) is an aggressive bone malignancy that mainly affects children and young adults. The mechanisms by which EWS (EWSR1) fusion genes drive the disease are not fully understood. CRM1 (XPO1) traffics proteins from the nucleus, including tumor suppressors and growth factors, and is overexpressed in many cancers. A small-molecule inhibitor of CRM1, KPT-330, has shown therapeutic promise, but has yet to be investigated in the context of EWS. In this study, we demonstrate that CRM1 is also highly expressed in EWS. shRNA-mediated or pharmacologic inhibition of CRM1 in EWS cells dramatically decreased cell growth while inducing apoptosis, cell-cycle arrest, and protein expression alterations to several cancer-related factors. Interestingly, silencing of CRM1 markedly reduced EWS-FLI1 fusion protein expression at the posttranscriptional level and upregulated the expression of the well-established EWS-FLI1 target gene, insulin-like growth factor binding protein 3 (IGFBP3), which inhibits IGF-1. Accordingly, KPT-330 treatment attenuated IGF-1-induced activation of the IGF-1R/AKT pathway. Furthermore, knockdown of IGFBP3 increased cell growth and rescued the inhibitory effects on IGF-1 signaling triggered by CRM1 inhibition. Finally, treatment of EWS cells with a combination of KPT-330 and the IGF-1R inhibitor, linsitinib, synergistically decreased cell proliferation both in vitro and in vivo Taken together, these findings provide a strong rationale for investigating the efficacy of combinatorial inhibition of CRM1 and IGF-1R for the treatment of EWS. Cancer Res; 76(9); 2687-97. ©2016 AACR.
Collapse
Affiliation(s)
- Haibo Sun
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, California. Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - De-Chen Lin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California. Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| | - Qi Cao
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Xiao Guo
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Helene Marijon
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Zhiqiang Zhao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Sigal Gery
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Liang Xu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Brendan Pang
- Department of Pathology, National University Hospital Singapore, Singapore
| | | | - Huey Jin Lim
- Department of Pathology, National University Hospital Singapore, Singapore
| | - Ngan Doan
- Department of Pathology and Laboratory Medicine, UCLA School of Medicine, Los Angeles, California
| | - Jonathan W Said
- Department of Pathology and Laboratory Medicine, UCLA School of Medicine, Los Angeles, California
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical Center, Los Angeles, California
| | - Anand Mayakonda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Tom Thomas
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Charles Forscher
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | | | | | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, California
| | - H Phillip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California. Cancer Science Institute of Singapore, National University of Singapore, Singapore. National University Cancer Institute, National University Hospital Singapore, Singapore
| |
Collapse
|
222
|
Kroiss M, Fassnacht M. Inhibition of Cholesterol Esterification in the Adrenal Gland by ATR101/PD132301-2, A Promising Case of Drug Repurposing. Endocrinology 2016; 157:1719-21. [PMID: 27149038 DOI: 10.1210/en.2016-1210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Matthias Kroiss
- Division of Endocrinology and Diabetology, Department of Internal Medicine, University Hospital Würzburg, University of Würzburg; and Clinical Chemistry and Laboratory Medicine, University Hospital Würzburg; and Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetology, Department of Internal Medicine, University Hospital Würzburg, University of Würzburg; and Clinical Chemistry and Laboratory Medicine, University Hospital Würzburg; and Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
223
|
Cheng Y, Kerppola RE, Kerppola TK. ATR-101 disrupts mitochondrial functions in adrenocortical carcinoma cells and in vivo. Endocr Relat Cancer 2016; 23:1-19. [PMID: 26843528 PMCID: PMC4887102 DOI: 10.1530/erc-15-0527] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/02/2016] [Indexed: 12/26/2022]
Abstract
Adrenocortical carcinoma (ACC) generally has poor prognosis. Existing treatments provide limited benefit for most patients with locally advanced or metastatic tumors. We investigated the mechanisms for the cytotoxicity, xenograft suppression, and adrenalytic activity of ATR-101 (PD132301-02), a prospective agent for ACC treatment. Oral administration of ATR-101 inhibited the establishment and impeded the growth of ACC-derived H295R cell xenografts in mice. ATR-101 induced H295R cell apoptosis in culture and in xenografts. ATR-101 caused mitochondrial hyperpolarization, reactive oxygen release, and ATP depletion within hours after exposure, followed by cytochrome c release, caspase-3/7 activation, and membrane permeabilization. The increase in mitochondrial membrane potential occurred concurrently with the decrease in cellular ATP levels. When combined with ATR-101, lipophilic free radical scavengers suppressed the reactive oxygen release, and glycolytic precursors prevented the ATP depletion, abrogating ATR-101 cytotoxicity. ATR-101 directly inhibited F1F0-ATPase activity and suppressed ATP synthesis in mitochondrial fractions. ATR-101 administration to guinea pigs caused oxidized lipofuscin accumulation in the zona fasciculate layer of the adrenal cortex, implicating reactive oxygen release in the adrenalytic effect of ATR-101. These results support the development of ATR-101 and other adrenalytic compounds for the treatment of ACC.
Collapse
Affiliation(s)
- Yunhui Cheng
- Department of Biological ChemistryUniversity of Michigan, Ann Arbor, MI, USA
| | | | - Tom Klaus Kerppola
- Department of Biological ChemistryUniversity of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
224
|
Schwartz GK, Dickson MA, LoRusso PM, Sausville EA, Maekawa Y, Watanabe Y, Kashima N, Nakashima D, Akinaga S. Preclinical and first-in-human phase I studies of KW-2450, an oral tyrosine kinase inhibitor with insulin-like growth factor receptor-1/insulin receptor selectivity. Cancer Sci 2016; 107:499-506. [PMID: 26850678 PMCID: PMC4832855 DOI: 10.1111/cas.12906] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/18/2016] [Accepted: 01/26/2016] [Indexed: 01/07/2023] Open
Abstract
Numerous solid tumors overexpress or have excessively activated insulin-like growth factor receptor-1 (IGF-1R). We summarize preclinical studies and the first-in-human study of KW-2450, an oral tyrosine kinase inhibitor with IGF-1R and insulin receptor (IR) inhibitory activity. Preclinical activity of KW-2450 was evaluated in various in vitro and in vivo models. It was then evaluated in a phase I clinical trial in 13 patients with advanced solid tumors (NCT00921336). In vitro, KW-2450 inhibited human IGF-1R and IR kinases (IC50 7.39 and 5.64 nmol/L, respectively) and the growth of various human malignant cell lines. KW-2450 40 mg/kg showed modest growth inhibitory activity and inhibited IGF-1-induced signal transduction in the murine HT-29/GFP colon carcinoma xenograft model. The maximum tolerated dose of KW-2450 was 37.5 mg once daily continuously; dose-limiting toxicity occurred in two of six patients at 50 mg/day (both grade 3 hyperglycemia) and in one of seven patients at 37.5 mg/day (grade 3 rash). Four of 10 evaluable patients showed stable disease. Single-agent KW-2450 was associated with modest antitumor activity in heavily pretreated patients with solid tumors and is being further investigated in combination therapy with lapatinib/letrozole in patients with human epidermal growth factor receptor 2-postive metastatic breast cancer.
Collapse
Affiliation(s)
- Gary K Schwartz
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, USA
| | - Mark A Dickson
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA, USA.,Weill Cornell Medical College, New York, NY, USA, USA
| | | | | | - Yoshimi Maekawa
- Fuji Research Park, R&D Division, Kyowa Hakko Kirin Co., Ltd., Shizuoka, Japan
| | - Yasuo Watanabe
- Fuji Research Park, R&D Division, Kyowa Hakko Kirin Co., Ltd., Shizuoka, Japan
| | - Naomi Kashima
- Fuji Research Park, R&D Division, Kyowa Hakko Kirin Co., Ltd., Shizuoka, Japan
| | - Daisuke Nakashima
- Planning Department, Kyowa Hakko Kirin Pharma Inc., Princeton, NJ, USA
| | - Shiro Akinaga
- Tokyo Research Park, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| |
Collapse
|
225
|
Hantel C, Ozimek A, Lira R, Ragazzon B, Jäckel C, Frantsev R, Reincke M, Bertherat J, Mussack T, Beuschlein F. TNF alpha signaling is associated with therapeutic responsiveness to vascular disrupting agents in endocrine tumors. Mol Cell Endocrinol 2016; 423:87-95. [PMID: 26768118 DOI: 10.1016/j.mce.2015.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 02/08/2023]
Abstract
ASA404 (Vadimezan) belongs to a class of agents with disrupting properties against tumor vasculature, which is partly mediated by TNFα-signaling. Preclinical and early clinical studies have indicated promising results for ASA404, while extended clinical trials performed poorly. Our aim was to investigate the potential therapeutic applicability of ASA404 against endocrine tumors. Moreover, as the reason for the unpredictable clinical anti-tumor activity of ASA 404 remained uncertain in previous studies, we compared two tumor models of endocrine origin with different responses to ASA404 treatment. Specifically, we determined anti-tumoral effects in preclinical models of neuroendocrine tumors of the gastroenteropancreatic system (BON) and adrenocortical cancer (NCI-H295R) in vitro and in xenograft models in vivo. Upon treatment of tumor bearing mice significant anti-tumoral effects, an increase in TNFα as well as activation of TNFα-specific downstream signaling were evident in the BON tumor model while no comparable effects were detectable for NCI-H295R. We identified TNFAIP3/A20, a key molecule of an inhibitory feedback-loop downstream of TNF-receptor 1, CD40, Toll-like receptors, NOD-like receptors and the interleukin-1 receptor signaling cascades, as overexpressed in the adrenocortical carcinoma tumor model. Subsequent analyses of clinical patient samples confirmed a correlation between tumor TNFAIP3 expression levels and overall survival in patients with ACC. Taken together our findings provide evidence that modulation of TNFα-signaling could be of relevance both for the clinical course of ACC patients and as a marker of treatment response.
Collapse
Affiliation(s)
- Constanze Hantel
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.
| | - Alexandra Ozimek
- Department of Surgery, Klinikum der Universität München, Munich, Germany
| | - Regia Lira
- Pediatrics Department, FMRP-USP, Ribeirão Preto, Brazil
| | - Bruno Ragazzon
- Institut Cochin, Inserm U1016, Cnrs UMR8104, Université Paris Descartes, Paris, France
| | - Carsten Jäckel
- Clinical Biochemistry, Medizinische Klinik und Poliklinik IV, Munich, Germany
| | - Roman Frantsev
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Martin Reincke
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Jérôme Bertherat
- Institut Cochin, Inserm U1016, Cnrs UMR8104, Université Paris Descartes, Paris, France
| | - Thomas Mussack
- Department of Surgery, Klinikum der Universität München, Munich, Germany
| | - Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
226
|
Bussey KJ, Bapat A, Linnehan C, Wandoloski M, Dastrup E, Rogers E, Gonzales P, Demeure MJ. Targeting polo-like kinase 1, a regulator of p53, in the treatment of adrenocortical carcinoma. Clin Transl Med 2016; 5:1. [PMID: 26754547 PMCID: PMC4709336 DOI: 10.1186/s40169-015-0080-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/15/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is an aggressive cancer with a 5 year survival rate of 20-30 %. Various factors have been implicated in the pathogenesis of ACC including dysregulation of the G2/M transition and aberrant activity of p53 and MDM2. Polo-like kinase 1 (PLK-1) negatively modulates p53 functioning, promotes MDM2 activity through its phosphorylation, and is involved in the G2/M transition. Gene expression profiling of 44 ACC samples showed that increased expression of PLK-1 in 29 % of ACC. Consequently, we examined PLK-1's role in the modulation of the p53 signaling pathway in adrenocortical cancer. METHODS We used siRNA knock down PLK-1 and pharmacological inhibition of PLK-1 and MDM2 ACC cell lines SW-13 and H295R. We examined viability, protein expression, p53 transactivation, and induction of apoptosis. RESULTS Knocking down expression of PLK-1 with siRNA or inhibition of PLK-1 by a small molecule inhibitor, BI-2536, resulted in a loss of viability of up to 70 % in the ACC cell lines H295R and SW-13. In xenograft models, BI-2536 demonstrated marked inhibition of growth of SW-13 with less inhibition of H295R. BI-2536 treatment resulted in a decrease in mutant p53 protein in SW-13 cells but had no effect on wild-type p53 protein levels in H295R cells. Additionally, inhibition of PLK-1 restored wild-type p53's transactivation and apoptotic functions in H295R cells, while these functions of mutant p53 were restored only to a smaller extent. Furthermore, inhibition of MDM2 with nutlin-3 reduced the viability of both the ACC cells and also reactivated wild-type p53's apoptotic function. Inhibition of PLK-1 sensitized the ACC cell lines to MDM2 inhibition and this dual inhibition resulted in an additive apoptotic response in H295R cells with wild-type p53. CONCLUSIONS These preclinical studies suggest that targeting p53 through PLK-1 is an attractive chemotherapy strategy warranting further investigation in adrenocortical cancer.
Collapse
Affiliation(s)
- Kimberly J Bussey
- NantOmics, LLC, The Biodesign Institute, Arizona State University, PO Box 875001, Tempe, AZ, 85287-5001, USA.
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Aditi Bapat
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Claire Linnehan
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | | | - Erica Dastrup
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Erik Rogers
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Paul Gonzales
- Translational Drug Development (TD2), Scottsdale, AZ, USA.
| | | |
Collapse
|
227
|
Massey PR, Wang R, Prasad V, Bates SE, Fojo T. Assessing the Eventual Publication of Clinical Trial Abstracts Submitted to a Large Annual Oncology Meeting. Oncologist 2016; 21:261-8. [PMID: 26888691 DOI: 10.1634/theoncologist.2015-0516] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Despite the ethical imperative to publish clinical trials when human subjects are involved, such data frequently remain unpublished. The objectives were to tabulate the rate and ascertain factors associated with eventual publication of clinical trial results reported as abstracts in the Proceedings of the American Society of Clinical Oncology (American Society of Clinical Oncology). MATERIALS AND METHODS Abstracts describing clinical trials for patients with breast, lung, colorectal, ovarian, and prostate cancer from 2009 to 2011 were identified by using a comprehensive online database (http://meetinglibrary.asco.org/abstracts). Abstracts included reported results of a treatment or intervention assessed in a discrete, prospective clinical trial. Publication status at 4-6 years was determined by using a standardized search of PubMed. Primary outcomes were the rate of publication for abstracts of randomized and nonrandomized clinical trials. Secondary outcomes included factors influencing the publication of results. RESULTS A total of 1,075 abstracts describing 378 randomized and 697 nonrandomized clinical trials were evaluated. Across all years, 75% of randomized and 54% of nonrandomized trials were published, with an overall publication rate of 61%. Sample size was a statistically significant predictor of publication for both randomized and nonrandomized trials (odds ratio [OR] per increase of 100 participants = 1.23 [1.11-1.36], p < .001; and 1.64 [1.15-2.34], p = .006, respectively). Among randomized studies, an industry coauthor or involvement of a cooperative group increased the likelihood of publication (OR 2.37, p = .013; and 2.21, p = .01, respectively). Among nonrandomized studies, phase II trials were more likely to be published than phase I (p < .001). Use of an experimental agent was not a predictor of publication in randomized (OR 0.76 [0.38-1.52]; p = .441) or nonrandomized trials (OR 0.89 [0.61-1.29]; p = .532). CONCLUSION This is the largest reported study examining why oncology trials are not published. The data show that 4-6 years after appearing as abstracts, 39% of oncology clinical trials remain unpublished. Larger sample size and advanced trial phase were associated with eventual publication; among randomized trials, an industry-affiliated author or a cooperative group increased likelihood of publication. Unfortunately, we found that, despite widespread recognition of the problem and the creation of central data repositories, timely publishing of oncology clinical trials results remains unsatisfactory.
Collapse
Affiliation(s)
- Paul R Massey
- Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Ruibin Wang
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vinay Prasad
- Medical Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Susan E Bates
- Medical Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tito Fojo
- Medical Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
228
|
Basham KJ, Hung HA, Lerario AM, Hammer GD. Mouse models of adrenocortical tumors. Mol Cell Endocrinol 2016; 421:82-97. [PMID: 26678830 PMCID: PMC4720156 DOI: 10.1016/j.mce.2015.11.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 12/17/2022]
Abstract
The molecular basis of the organogenesis, homeostasis, and tumorigenesis of the adrenal cortex has been the subject of intense study for many decades. Specifically, characterization of tumor predisposition syndromes with adrenocortical manifestations and molecular profiling of sporadic adrenocortical tumors have led to the discovery of key molecular pathways that promote pathological adrenal growth. However, given the observational nature of such studies, several important questions regarding the molecular pathogenesis of adrenocortical tumors have remained. This review will summarize naturally occurring and genetically engineered mouse models that have provided novel tools to explore the molecular and cellular underpinnings of adrenocortical tumors. New paradigms of cancer initiation, maintenance, and progression that have emerged from this work will be discussed.
Collapse
Affiliation(s)
- Kaitlin J Basham
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, USA; Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Holly A Hung
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, USA; Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, USA; Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, USA; Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
229
|
Lalli E, Sasano H. 5th International ACC Symposium: An Outlook to Current and Future Research on the Biology of Adrenocortical Carcinoma: Diagnostic and Therapeutic Applications. HORMONES & CANCER 2016; 7:44-48. [PMID: 26666256 PMCID: PMC10355864 DOI: 10.1007/s12672-015-0240-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/01/2015] [Indexed: 12/27/2022]
Abstract
Groundbreaking progress has been recently made in elucidating the signaling pathways that are altered in adrenocortical carcinoma (ACC), an endocrine malignancy that still has an unfavorable prognosis, and in understanding its genomic structure. These advances need now to be translated to create cellular and animal models more relevant to human disease in order to develop new and more effective diagnostic procedures and targeted therapies against this deadly malignancy.
Collapse
Affiliation(s)
- Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, 660 route des Lucioles, 06560, Valbonne, France.
- NEOGENEX CNRS International Associated Laboratory, Valbonne, France.
- Université de Nice-Sophia Antipolis, Nice, France.
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku Sendai, 980-8575, Japan.
| |
Collapse
|
230
|
Macaulay VM, Middleton MR, Eckhardt SG, Rudin CM, Juergens RA, Gedrich R, Gogov S, McCarthy S, Poondru S, Stephens AW, Gadgeel SM. Phase I Dose-Escalation Study of Linsitinib (OSI-906) and Erlotinib in Patients with Advanced Solid Tumors. Clin Cancer Res 2016; 22:2897-907. [PMID: 26831715 DOI: 10.1158/1078-0432.ccr-15-2218] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/02/2016] [Indexed: 01/08/2023]
Abstract
PURPOSE Cross-talk between type I IGF receptor (IGF1R), insulin receptor (INSR), and epidermal growth factor receptor (EGFR) mediates resistance to individual receptor blockade. This study aimed to determine the MTD, safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of linsitinib, a potent oral IGF1R/INSR inhibitor, with EGFR inhibitor erlotinib. EXPERIMENTAL DESIGN This open-label, dose-escalation study investigated linsitinib schedules S1: once daily intermittent (days 1-3 weekly); S2, once daily continuous; S3, twice-daily continuous; each with erlotinib 100-150 mg once daily; and a non-small cell lung cancer (NSCLC) expansion cohort. RESULTS Ninety-five patients were enrolled (S1, 44; S2, 24; S3, 12; expansion cohort, 15) and 91 treated. Seven experienced dose-limiting toxicities: QTc prolongation (3), abnormal liver function (2), hyperglycemia (1), and anorexia (1). Common adverse events included drug eruption (84%), diarrhea (73%), fatigue (68%), nausea (58%), vomiting (40%). MTDs for linsitinib/erlotinib were 450/150 mg (S1), 400/100 mg (S2). On the basis of prior monotherapy data, S3 dosing at 150 mg twice daily/150 mg once daily was the recommended phase II dose for the expansion cohort. There was no evidence of drug-drug interaction. Pharmacodynamic data showed IGF-1 elevation and reduced IGF1R/INSR phosphorylation, suggesting pathway inhibition. Across schedules, 5/75 (7%) evaluable patients experienced partial responses: spinal chordoma (268+ weeks), rectal cancer (36 weeks), three NSCLCs including 2 adenocarcinomas (16, 72 weeks), 1 squamous wild-type EGFR NSCLC (36 weeks). Disease control (CR+PR+SD) occurred in 38 of 75 (51%), and 28 of 91 (31%) patients were on study >12 weeks. CONCLUSIONS The linsitinib/erlotinib combination was tolerable with preliminary evidence of activity, including durable responses in cases unlikely to respond to erlotinib monotherapy. Clin Cancer Res; 22(12); 2897-907. ©2016 AACR.
Collapse
Affiliation(s)
- Valentine M Macaulay
- University Department of Oncology, Oxford Cancer and Haematology Centre, Headington, Oxford, United Kingdom.
| | - Mark R Middleton
- University Department of Oncology, Oxford Cancer and Haematology Centre, Headington, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Bertoni N, Pereira LMS, Severino FE, Moura R, Yoshida WB, Reis PP. Integrative meta-analysis identifies microRNA-regulated networks in infantile hemangioma. BMC MEDICAL GENETICS 2016; 17:4. [PMID: 26772808 PMCID: PMC4715339 DOI: 10.1186/s12881-015-0262-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/12/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Hemangioma is a common benign tumor in the childhood; however our knowledge about the molecular mechanisms of hemangioma development and progression are still limited. Currently, microRNAs (miRNAs) have been shown as gene expression regulators with an important role in disease pathogenesis. Our goals were to identify miRNA-mRNA expression networks associated with infantile hemangioma. METHODS We performed a meta-analysis of previously published gene expression datasets including 98 hemangioma samples. Deregulated genes were further used to identify microRNAs as potential regulators of gene expression in infantile hemangioma. Data were integrated using bioinformatics methods, and genes were mapped in proteins, which were then used to construct protein-protein interaction networks. RESULTS Deregulated genes play roles in cell growth and differentiation, cell signaling, angiogenesis and vasculogenesis. Regulatory networks identified included microRNAs miR-9, miR-939 and let-7 family; these microRNAs showed the most number of interactions with deregulated genes in infantile hemangioma, suggesting that they may have an important role in the molecular mechanisms of disease. Additionally, results were used to identify drug-gene interactions and druggable gene categories using Drug-Gene Interaction Database. We show that microRNAs and microRNA-target genes may be useful biomarkers for the development of novel therapeutic strategies for patients with infantile hemangioma. CONCLUSIONS microRNA-regulated pathways may play a role in infantile hemangioma development and progression and may be potentially useful for future development of novel therapeutic strategies for patients with infantile hemangioma.
Collapse
Affiliation(s)
- Natália Bertoni
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University-UNESP, Av. Prof. Montenegro, 18618-970, Botucatu, São Paulo, Brazil.
| | - Lied M S Pereira
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University-UNESP, Av. Prof. Montenegro, 18618-970, Botucatu, São Paulo, Brazil.
| | - Fábio E Severino
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University-UNESP, Av. Prof. Montenegro, 18618-970, Botucatu, São Paulo, Brazil.
| | - Regina Moura
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University-UNESP, Av. Prof. Montenegro, 18618-970, Botucatu, São Paulo, Brazil.
| | - Winston B Yoshida
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University-UNESP, Av. Prof. Montenegro, 18618-970, Botucatu, São Paulo, Brazil.
| | - Patricia P Reis
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University-UNESP, Av. Prof. Montenegro, 18618-970, Botucatu, São Paulo, Brazil.
| |
Collapse
|
232
|
5th International ACC Symposium: Future and Current Therapeutic Trials in Adrenocortical Carcinoma. Discov Oncol 2016; 7:29-35. [PMID: 26728470 DOI: 10.1007/s12672-015-0241-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare and complex disease associated with a high mortality rate. Despite intensive translational and clinical research, prognosis remains poor. Over the past decade, a significant effort has been made to develop multinational, collaborative studies to better understand the pathogenesis and clinical features of this rare disease in attempt to improve the therapeutic strategies and patient outcome. The results of both standard and newer treatments are discussed in this review as well as the recent discovery of pathways involved in ACC pathogenesis that provide the rationale to introduce new molecular target therapies. Finally, remaining issues regarding how to improve available therapies in adjuvant setting are raised and addressed.
Collapse
|
233
|
Stigliano A, Chiodini I, Giordano R, Faggiano A, Canu L, Della Casa S, Loli P, Luconi M, Mantero F, Terzolo M. Management of adrenocortical carcinoma: a consensus statement of the Italian Society of Endocrinology (SIE). J Endocrinol Invest 2016; 39:103-21. [PMID: 26165270 DOI: 10.1007/s40618-015-0349-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/23/2015] [Indexed: 01/10/2023]
Affiliation(s)
- A Stigliano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | - I Chiodini
- Endocrinology and Metabolic Disease Unit, IRCCS Foundation Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - R Giordano
- Department of Clinical and Biological Science, University of Turin, Turin, Italy
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Science, University of Turin, Turin, Italy
| | - A Faggiano
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - L Canu
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - S Della Casa
- Endocrinology, Department of Internal Medicine, Catholic University of Rome, Rome, Italy
| | - P Loli
- Endocrine Unit, Niguarda Cà Granda Hospital, Milan, Italy
| | - M Luconi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - F Mantero
- Endocrinology Unit, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - M Terzolo
- Internal Medicine I, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
234
|
Cabezon-Gutierrez L, Franco-Moreno AI, Khosravi-Shahi P, Custodio-Cabello S, Garcia-Navarro MJ, Martin-Diaz RM. Clinical Case of Metastatic Adrenocortical Carcinoma With Unusual Evolution: Review the Literature. World J Oncol 2015; 6:485-490. [PMID: 28983351 PMCID: PMC5624676 DOI: 10.14740/wjon936w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2015] [Indexed: 12/13/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare and heterogeneous malignancy, with an incidence of approximately 0.72 per million cases per year leading to 0.2% of all cancer deaths in the United States. Metastatic ACC has a dismal prognosis with an overall survival of less than 1 year. We present a case of a 37-year-old man with metastatic ACC with unusual good prognosis and review the therapeutic options in the literature.
Collapse
|
235
|
Altieri B, Tirabassi G, Della Casa S, Ronchi CL, Balercia G, Orio F, Pontecorvi A, Colao A, Muscogiuri G. Adrenocortical tumors and insulin resistance: What is the first step? Int J Cancer 2015; 138:2785-94. [PMID: 26637955 DOI: 10.1002/ijc.29950] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/05/2015] [Accepted: 11/23/2015] [Indexed: 01/15/2023]
Abstract
The pathogenetic mechanisms underlying the onset of adrenocortical tumors (ACTs) are still largely unknown. Recently, more attention has been paid to the role of insulin and insulin-like growth factor (IGF) system on general tumor development and progression. Increased levels of insulin, IGF-1 and IGF-2 are associated with tumor cell growth and increased risk of cancer promotion and progression in patients with type 2 diabetes. Insulin resistance and compensatory hyperinsulinemia may play a role in adrenal tumor growth through the activation of insulin and IGF-1 receptors. Interestingly, apparently non-functioning ACTs are often associated with a high prevalence of insulin resistance and metabolic syndrome. However, it is unclear if ACT develops from a primary insulin resistance and compensatory hyperinsulinemia or if insulin resistance is only secondary to the slight cortisol hypersecretion by ACT. The aim of this review is to summarize the current evidence regarding the relationship between hyperinsulinemia and adrenocortical tumors.
Collapse
Affiliation(s)
- Barbara Altieri
- Institute of Medical Pathology, Division of Endocrinology and Metabolic Diseases, Catholic University, Rome, Italy
| | - Giacomo Tirabassi
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Silvia Della Casa
- Institute of Medical Pathology, Division of Endocrinology and Metabolic Diseases, Catholic University, Rome, Italy
| | - Cristina L Ronchi
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Giancarlo Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Orio
- Department of Sports Science and Wellness, Parthenope University, Naples, Italy.,Department of Endocrinology and Diabetology, Fertility Techniques Structure, University Hospital S. Giovanni Di Dio E Ruggi D'aragona, Salerno, Italy
| | - Alfredo Pontecorvi
- Institute of Medical Pathology, Division of Endocrinology and Metabolic Diseases, Catholic University, Rome, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Section of Endocrinology, Federico II University, Naples, Italy
| | - Giovanna Muscogiuri
- Department of Clinical Medicine and Surgery, Section of Endocrinology, Federico II University, Naples, Italy
| |
Collapse
|
236
|
Yang HA, Wang X, Ding F, Pang Q. MiRNA-323-5p Promotes U373 Cell Apoptosis by Reducing IGF-1R. Med Sci Monit 2015; 21:3880-6. [PMID: 26656446 PMCID: PMC4681375 DOI: 10.12659/msm.895037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/28/2015] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND MicroRNA regulates mammalian cell growth in terms of its proliferation and apoptosis by controlling the expression of target genes. MiRNA-323-5p plays an important role in regulating cell growth and death within various types of cells. The function of miRNA-323-5p and its possible molecular mechanism in human cerebral glioma U373 cells remains to be further confirmed. The aim of this study was to investigate the regulation function of miRNA-323-5p in human glioma U373 cell growth, proliferation, and apoptosis. MATERIAL AND METHODS We used human cerebral glioma U373 cells as the cell model; utilized liposome technology (transfected by Lipofectamine2000) in human cerebral glioma U373 cells to over-express miRNA-323-5p (microRNA used as control group); and selected MTT assay and flow cytometry to detect cell growth, proliferation, and apoptosis. We used RT-PCR and Western blotting techniques to study the expression levels of target insulin-like growth factor 1 (IGF-1) receptor protein in U373 cells transfected with miRNA-323-5p. We used liposome transfection techniques in human cerebral glioma U373 cells to over-express or processed knockdown of IGF-1R by siRNA, and then transferred with miRNA-323-5p, thereby investigating the treated human cerebral glioma U373 cells apoptosis situations. RESULTS The over-expression of miRNA-323-5p inhibited the growth and proliferation of human cerebral glioma U373 cells and promoted its apoptosis. The over-expression of miRNA-323-5p also reduced the IGF-1R level. After processing the knockdown of IGF-1R and then transfection with miRNA-323-5p, U373 cells had enhanced apoptosis. The over-expression of IGF-1R inhibited the cells apoptosis induced by miRNA-323-5p. CONCLUSIONS MiRNA-323-5p inhibited human cerebral glioma U373 cell proliferation and promoted its apoptosis by reducing IGF-1R.
Collapse
Affiliation(s)
- Hong-an Yang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, , P.R. China
| | - Xiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Feng Ding
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, , P.R. China
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, , P.R. China
| |
Collapse
|
237
|
Kerkhofs TMA, Ettaieb MHT, Hermsen IGC, Haak HR. Developing treatment for adrenocortical carcinoma. Endocr Relat Cancer 2015; 22:R325-38. [PMID: 26259571 DOI: 10.1530/erc-15-0318] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2015] [Indexed: 02/02/2023]
Abstract
Cancer of the adrenal cortex (ACC) is a rare endocrine malignancy with limited treatment options. Patients typically present with autonomous hormonal overproduction and/or a large abdominal mass. Hormonal assays and medical imaging can be diagnostic, but urinary steroid profiling might be a more sensitive technique to assess malignancy in adrenal tumours. The stage of the disease at diagnosis is the most important prognostic factor. The current staging system needs refinement, especially to separate aggressive from indolent disease in stage IV patients and to select patients who need adjuvant treatment after complete surgical resection. Regarding the latter, assessing the proliferation index Ki-67 seems the best tool currently available. Genomic profiling is expected to become of clinical relevance in the future. Medical therapy is centred on the adrenolytic drug mitotane, which carries considerable toxicity and is not easy to manage. Its tolerability and long plasma level build-up phase may be improved by therapeutic drug monitoring based on pharmacokinetic modelling and intensive counselling of patients. Current chemotherapy regimens can offer disease stabilization in about 50% of patients, but an objective response should be expected in <25%. Research on targeted therapy and immunotherapy is difficult in this rare disease with often heavily pre-treated patients and has not yet been successful. Quality of care should be ensured by treating patients in centres with established experience in multidisciplinary oncologic care, who adhere to prevailing guidelines and state-of-the-art in diagnostic and treatment concepts. International collaboration in fundamental research and clinical trials is the key to further elucidate the pathogenesis and to improve patient care.
Collapse
Affiliation(s)
- T M A Kerkhofs
- Department of Internal MedicineMaxima Medical Center, Ds. Th. Fliednerstraat 1, 5631 BM Eindhoven/Veldhoven, The NetherlandsDivision of General Internal MedicineDepartment of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The NetherlandsDepartment of Health Services Research and CAPHRI School for Public Health and Primary CareMaastricht University Medical Center, The Netherlands
| | - M H T Ettaieb
- Department of Internal MedicineMaxima Medical Center, Ds. Th. Fliednerstraat 1, 5631 BM Eindhoven/Veldhoven, The NetherlandsDivision of General Internal MedicineDepartment of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The NetherlandsDepartment of Health Services Research and CAPHRI School for Public Health and Primary CareMaastricht University Medical Center, The Netherlands
| | - I G C Hermsen
- Department of Internal MedicineMaxima Medical Center, Ds. Th. Fliednerstraat 1, 5631 BM Eindhoven/Veldhoven, The NetherlandsDivision of General Internal MedicineDepartment of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The NetherlandsDepartment of Health Services Research and CAPHRI School for Public Health and Primary CareMaastricht University Medical Center, The Netherlands
| | - H R Haak
- Department of Internal MedicineMaxima Medical Center, Ds. Th. Fliednerstraat 1, 5631 BM Eindhoven/Veldhoven, The NetherlandsDivision of General Internal MedicineDepartment of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The NetherlandsDepartment of Health Services Research and CAPHRI School for Public Health and Primary CareMaastricht University Medical Center, The Netherlands Department of Internal MedicineMaxima Medical Center, Ds. Th. Fliednerstraat 1, 5631 BM Eindhoven/Veldhoven, The NetherlandsDivision of General Internal MedicineDepartment of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The NetherlandsDepartment of Health Services Research and CAPHRI School for Public Health and Primary CareMaastricht University Medical Center, The Netherlands Department of Internal MedicineMaxima Medical Center, Ds. Th. Fliednerstraat 1, 5631 BM Eindhoven/Veldhoven, The NetherlandsDivision of General Internal MedicineDepartment of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The NetherlandsDepartment of Health Services Research and CAPHRI School for Public Health and Primary CareMaastricht University Medical Center, The Netherlands
| |
Collapse
|
238
|
Abstract
Preclinical studies in the 1980s defined a role for IGF signaling in the development and sustainability of the malignant process. Subsequently, antibody, tyrosine kinase, and ligand inhibitors of the IGF receptor were manufactured. In the past decade, numerous clinical trials have tested the efficacy of IGF receptor inhibitors in the treatment of advanced tumors. Early-phase trials in heavily pretreated populations showed promise with complete or partial responses in a few patients and stable disease in many more. Unfortunately, the results of the early-phase trials did not pan out to later-phase trials. The lack of use of biomarkers to define subsets of patients that may benefit from IGF receptor blockade and compensatory signaling via other growth factor receptors such as the insulin, GH, and epidermal growth factor receptors may have played a role in the lack of efficacy of IGF receptor inhibition in phase III trials. Although these trials failed to show benefit, the trials have revealed previously unknown knowledge regarding the complex nature of IGF signaling. The knowledge obtained from these trials will be useful in designing future trials studying inhibitors of growth factor signaling.
Collapse
Affiliation(s)
- Heather Beckwith
- Departments of Medicine (H.B., D.Y.) and Pharmacology (D.Y.) and Masonic Cancer Center (D.Y.), University of Minnesota, Minneapolis, Minnesota 55455
| | - Douglas Yee
- Departments of Medicine (H.B., D.Y.) and Pharmacology (D.Y.) and Masonic Cancer Center (D.Y.), University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
239
|
Heidegger I, Massoner P, Sampson N, Klocker H. The insulin-like growth factor (IGF) axis as an anticancer target in prostate cancer. Cancer Lett 2015; 367:113-21. [PMID: 26231734 DOI: 10.1016/j.canlet.2015.07.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/18/2015] [Accepted: 07/21/2015] [Indexed: 12/21/2022]
Abstract
Prostate cancer (PCa) is the most common cancer and the second leading cause of cancer death in males. In recent years, several new targeting agents have been introduced for the treatment of advanced stages of the disease. However, development of resistance limits the efficacy of new drugs and there is a further need to develop additional novel treatment approaches. One of the most investigated targets in cancer research is the insulin-like growth factor (IGF) axis, whose receptors are overexpressed in several cancer entities including PCa. In preclinical studies in PCa, targeting of the IGF axis receptors showed promising anti-tumor effects. Currently available data on clinical studies do not meet the expectations for this new treatment approach. In this review we provide a summary of preclinical and clinical studies on the IGF axis in PCa including treatment with monoclonal antibodies and tyrosine kinase inhibitors. Moreover, we summarize preliminary results from ongoing studies and discuss limitations and side effects of the substances used. We also address the role of the IGF axis in the biomarkers setting including IGF-binding proteins and genetic variants.
Collapse
Affiliation(s)
- Isabel Heidegger
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Petra Massoner
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Natalie Sampson
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Helmut Klocker
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria.
| |
Collapse
|
240
|
Libé R. Adrenocortical carcinoma (ACC): diagnosis, prognosis, and treatment. Front Cell Dev Biol 2015; 3:45. [PMID: 26191527 PMCID: PMC4490795 DOI: 10.3389/fcell.2015.00045] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/22/2015] [Indexed: 12/16/2022] Open
Abstract
Adrenocortical carticnoma (ACC) is a rare malignancy with an incidence of 0.7-2.0 cases/million habitants/year. The diagnosis of malignancy relies on careful investigations of clinical, biological, and imaging features before surgery and pathological examination after tumor removal. Most patients present with steroid hormone excess or abdominal mass effects, but 15% of patients with ACC is initially diagnosed incidentally. After the diagnosis, in order to assess the ACC prognosis and establish an adequate basis for treatment decisions different tools are proposed. The stage classification proposed by the European Network for the Study of Adrenal Tumors (ENSAT) is recommended. Pathology reports define the Weiss score, the resection status and the proliferative index, including the mitotic count and the Ki67 index. As far as the treatment is concerned, in case of tumor limited to the adrenal gland, the complete resection of the tumor is the first option. Most patients benefit from adjuvant mitotane treatment. In metastatic disease, mitotane is the cornerstone of initial treatment, and cytotoxic drugs should be added in case of progression. Recently, the First International Randomized (FIRM-ACT) Trial in metastatic ACC reported the association between mitotane and etoposide/doxorubicin/cisplatin (EDP) as the new standard in first line treatment of ACC. In last years, new targeted therapies, including the IGF-1 receptor inhibitors, have been investigated, but their efficacy remains limited. Thus, new treatment concepts are urgently needed. The ongoing "omic approaches" and next-generation sequencing will improve our understanding of the pathogenesis and hopefully will lead to better therapies.
Collapse
Affiliation(s)
- Rossella Libé
- Department of Endocrinology, French Network for Adrenal Cancer, Cochin Hospital Paris, France
| |
Collapse
|
241
|
Ferrari L, Claps M, Grisanti S, Berruti A. Systemic Therapy in Locally Advanced or Metastatic Adrenal Cancers: A Critical Appraisal and Clinical Trial Update. Eur Urol Focus 2015; 1:298-300. [PMID: 28723406 DOI: 10.1016/j.euf.2015.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 05/16/2015] [Accepted: 06/07/2015] [Indexed: 11/26/2022]
Abstract
Mitotane and chemotherapy with etoposide, doxorubicin, and cisplatin plus mitotane (EDP-M) are the only therapies with demonstrated efficacy in advanced adrenocortical carcinoma. Prognostic and predictive factors are needed to identify patients who could obtain the best benefit from these treatments. Despite the strong rationale for their use, clinical trials on molecular targeted therapies have failed to demonstrate that these drugs are efficacious in the management of this extremely rare disease.
Collapse
Affiliation(s)
- Laura Ferrari
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Spedali Civili Hospital, Brescia, Italy
| | - Mélanie Claps
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Spedali Civili Hospital, Brescia, Italy
| | - Salvatore Grisanti
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Spedali Civili Hospital, Brescia, Italy
| | - Alfredo Berruti
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Spedali Civili Hospital, Brescia, Italy.
| |
Collapse
|
242
|
Abstract
Recent developments in the treatment of adrenocortical carcinoma (ACC) include diagnostic and prognostic risk stratification algorithms, increasing evidence of the impact of historical therapies on overall survival, and emerging targets from integrated epigenomic and genomic analyses. Advances include proper clinical and molecular characterization of all patients with ACC, standardization of proliferative index analyses, referral of these patients to large cancer referral centers at the time of first surgery, and development of new trials in patients with well-characterized ACC. Networking and progress in the molecular characterization of ACC constitute the basis for significant future therapeutic breakthroughs.
Collapse
Affiliation(s)
- Eric Baudin
- Département de Médecine, Gustave Roussy, 114, rue Édouard-Vaillant, Paris South University, Villejuif Cedex 94805, France; Département de Nucléaire et de Cancérologie Endocrinienne, Gustave Roussy, 114, rue Édouard-Vaillant, Paris South University, Villejuif Cedex 94805, France; Faculté de Médecine, INSERM UMR 1185, 63 rue Gabriel Péri, F-94276 Le Kremlin-Bicêtre, Université Paris Sud, Paris, France.
| |
Collapse
|
243
|
Puzanov I, Hess A. LK or IGF1R: When selectivity hurts. Aging (Albany NY) 2015; 7:342-3. [PMID: 26080834 PMCID: PMC4505153 DOI: 10.18632/aging.100753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Igor Puzanov
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | |
Collapse
|
244
|
|
245
|
Cherradi N. microRNAs as Potential Biomarkers in Adrenocortical Cancer: Progress and Challenges. Front Endocrinol (Lausanne) 2015; 6:195. [PMID: 26834703 PMCID: PMC4719100 DOI: 10.3389/fendo.2015.00195] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/27/2015] [Indexed: 12/11/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with poor prognosis and limited therapeutic options. Over the last decade, pan-genomic analyses of genetic and epigenetic alterations and genome-wide expression profile studies allowed major advances in the understanding of the molecular genetics of ACC. Besides the well-known dysfunctional molecular pathways in adrenocortical tumors, such as the IGF2 pathway, the Wnt pathway, and TP53, high-throughput technologies enabled a more comprehensive genomic characterization of adrenocortical cancer. Integration of expression profile data with exome sequencing, SNP array analysis, methylation, and microRNA (miRNA) profiling led to the identification of subgroups of malignant tumors with distinct molecular alterations and clinical outcomes. miRNAs post-transcriptionally silence their target gene expression either by degrading mRNA or by inhibiting translation. Although our knowledge of the contribution of deregulated miRNAs to the pathogenesis of ACC is still in its infancy, recent studies support their relevance in gene expression alterations in these tumors. Some miRNAs have been shown to carry potential diagnostic and prognostic values, while others may be good candidates for therapeutic interventions. With the emergence of disease-specific blood-borne miRNAs signatures, analyses of small cohorts of patients with ACC suggest that circulating miRNAs represent promising non-invasive biomarkers of malignancy or recurrence. However, some technical challenges still remain, and most of the miRNAs reported in the literature have not yet been validated in sufficiently powered and longitudinal studies. In this review, we discuss the current knowledge regarding the deregulation of tumor-associated and circulating miRNAs in ACC patients, while emphasizing their potential significance in pathogenic pathways in light of recent insights into the role of miRNAs in shaping the tumor microenvironment.
Collapse
Affiliation(s)
- Nadia Cherradi
- U1036, Institut National de la Santé et de la Recherche Médicale, Grenoble, France
- Biologie du Cancer et de l’Infection, Commissariat à l’Energie Atomique, Institut de Recherches en Technologies et Sciences pour le Vivant, Grenoble, France
- Laboratoire BCI, Université Grenoble-Alpes, Grenoble, France
- *Correspondence: Nadia Cherradi,
| |
Collapse
|
246
|
Robbins HL, Hague A. The PI3K/Akt Pathway in Tumors of Endocrine Tissues. Front Endocrinol (Lausanne) 2015; 6:188. [PMID: 26793165 PMCID: PMC4707207 DOI: 10.3389/fendo.2015.00188] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/07/2015] [Indexed: 12/29/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a key driver in carcinogenesis. Defects in this pathway in human cancer syndromes such as Cowden's disease and Multiple Endocrine Neoplasia result in tumors of endocrine tissues, highlighting its importance in these cancer types. This review explores the growing evidence from multiple animal and in vitro models and from analysis of human tumors for the involvement of this pathway in the following: thyroid carcinoma subtypes, parathyroid carcinoma, pituitary tumors, adrenocortical carcinoma, phaeochromocytoma, neuroblastoma, and gastroenteropancreatic neuroendocrine tumors. While data are not always consistent, immunohistochemistry performed on human tumor tissue has been used alongside other techniques to demonstrate Akt overactivation. We review active Akt as a potential prognostic marker and the PI3K pathway as a therapeutic target in endocrine neoplasia.
Collapse
Affiliation(s)
- Helen Louise Robbins
- Department of General Surgery, University Hospital Coventry and Warwickshire, Coventry, UK
| | - Angela Hague
- School of Oral and Dental Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- *Correspondence: Angela Hague,
| |
Collapse
|